1
|
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
|
2
|
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
|
3
|
Abrigo J, Simon F, Cabrera D, Vilos C, Cabello-Verrugio C. Combined Administration of Andrographolide and Angiotensin- (1-7) Synergically Increases the Muscle Function and Strength in Aged Mice. Curr Mol Med 2021; 22:908-918. [PMID: 34875988 DOI: 10.2174/1566524021666211207112106] [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: 12/25/2020] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Sarcopenia is a progressive and generalized skeletal muscle disorder characterized by muscle weakness, loss of muscle mass, and decline in the capacity of force generation. Aging can cause sarcopenia. Several therapeutic strategies have been evaluated to prevent or alleviate this disorder. One of them is angiotensin 1-7 [Ang-(1-7)], an anti-atrophic peptide for skeletal muscles that regulates decreased muscle mass for several causes, including aging. Another regulator of muscle mass and function is andrographolide, a bicyclic diterpenoid lactone that decreases the nuclear factor kappa B (NF-κB) signaling and attenuates the severity of some muscle diseases. OBJECTIVE Evaluate the effect of combined administration of Ang-(1-7) with andrographolide on the physical performance, muscle strength, and fiber´s diameter in a murine model of sarcopenia by aging. METHODS Aged male mice of the C57BL/6J strain were treated with Andrographolide, Ang-(1-7), or combined for three months. The physical performance, muscle strength, and fiber´s diameter were measured. RESULTS The results showed that aged mice (24 months old) treated with Ang-(1-7) or Andrographolide improved their performance on a treadmill test, muscle strength, and their fiber´s diameter compared to aged mice without treatment. The combined administration of Ang-(1-7) with andrographolide to aged mice has an enhanced synergically effect on physical performance, muscle strength, and fiber´s diameter. CONCLUSION Our results indicated that in aged mice, the effects of andrographolide and Ang-(1-7) on muscle function, strength, and fiber´s diameter are potentiated.
Collapse
Affiliation(s)
- Johanna Abrigo
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello. Santiago. Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy. Santiago. Chile
| | - Daniel Cabrera
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile. Santiago. Chile
| | - Cristian Vilos
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile. Santiago. Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello. Santiago. Chile
| |
Collapse
|
4
|
Panza E, Vellecco V, Iannotti FA, Paris D, Manzo OL, Smimmo M, Mitilini N, Boscaino A, de Dominicis G, Bucci M, Di Lorenzo A, Cirino G. Duchenne's muscular dystrophy involves a defective transsulfuration pathway activity. Redox Biol 2021; 45:102040. [PMID: 34174560 PMCID: PMC8246642 DOI: 10.1016/j.redox.2021.102040] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/20/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most frequent X chromosome-linked disease caused by mutations in the gene encoding for dystrophin, leading to progressive and unstoppable degeneration of skeletal muscle tissues. Despite recent advances in the understanding of the molecular processes involved in the pathogenesis of DMD, there is still no cure. In this study, we aim at investigating the potential involvement of the transsulfuration pathway (TSP), and its by-end product namely hydrogen sulfide (H2S), in primary human myoblasts isolated from DMD donors and skeletal muscles of dystrophic (mdx) mice. In myoblasts of DMD donors, we demonstrate that the expression of key genes regulating the H2S production and TSP activity, including cystathionine γ lyase (CSE), cystathionine beta-synthase (CBS), 3 mercaptopyruvate sulfurtransferase (3-MST), cysteine dioxygenase (CDO), cysteine sulfonic acid decarboxylase (CSAD), glutathione synthase (GS) and γ -glutamylcysteine synthetase (γ-GCS) is reduced. Starting from these findings, using Nuclear Magnetic Resonance (NMR) and quantitative Polymerase Chain Reaction (qPCR) we show that the levels of TSP-related metabolites such as methionine, glycine, glutathione, glutamate and taurine, as well as the expression levels of the aforementioned TSP related genes, are significantly reduced in skeletal muscles of mdx mice compared to healthy controls, at both an early (7 weeks) and overt (17 weeks) stage of the disease. Importantly, the treatment with sodium hydrosulfide (NaHS), a commonly used H2S donor, fully recovers the impaired locomotor activity in both 7 and 17 old mdx mice. This is an effect attributable to the reduced expression of pro-inflammatory markers and restoration of autophagy in skeletal muscle tissues. In conclusion, our study uncovers a defective TSP pathway activity in DMD and highlights the role of H2S-donors for novel and safe adjuvant therapy to treat symptoms of DMD.
Collapse
Affiliation(s)
- E Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - V Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - F A Iannotti
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli (NA), Italy
| | - D Paris
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli (NA), Italy
| | - O L Manzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy; Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - M Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - N Mitilini
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - A Boscaino
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - G de Dominicis
- UOSC, Pathological Anatomy, A. Cardarelli Hospital, Naples, Italy
| | - M Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - A Di Lorenzo
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - G Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Safety and clinical outcome of tamoxifen in Duchenne Muscular Dystrophy. Neuromuscul Disord 2021; 31:803-813. [PMID: 34304968 DOI: 10.1016/j.nmd.2021.05.005] [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: 12/24/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 01/25/2023]
Abstract
Patients having Duchenne Muscular Dystrophy (DMD) are currently being treated with corticosteroids, which slow down disease progression at the expense of serious adverse effects. Tamoxifen is a pro-drug some of whose metabolites interact with the nuclear estrogen receptor, leading to anti-fibrotic and muscle-protective effects as has been demonstrated in a murine model of DMD. Here we report the results from a monocentric single arm prospective study in 13 ambulant boys aged 6-14 years with genetically confirmed DMD, aimed to assess the safety of tamoxifen and its impact on disease progression. Boys were treated for up to 3 years with 20 mg/day of oral tamoxifen, in addition to their ongoing corticosteroid treatment. For 8 of these patients, outcome was compared to an age- and performance-matched 12-month natural history dataset. The primary end point was the 6-minute walk test. Secondary end points were the NorthStar assessment, timed function tests, pulmonary function, the biomarker creatine phosphokinase and adverse effects. No adverse effects were noticed other than mild gynecomastia in 4 boys. Tamoxifen-treated patients retained motor and respiratory function, compared with a significant deterioration of age-matched historical control patients receiving corticosteroids only. These encouraging findings warrant a larger clinical trial to substantiate the use of tamoxifen in Duchenne Muscular Dystrophy.
Collapse
|
6
|
Nogami K, Maruyama Y, Sakai-Takemura F, Motohashi N, Elhussieny A, Imamura M, Miyashita S, Ogawa M, Noguchi S, Tamura Y, Kira JI, Aoki Y, Takeda S, Miyagoe-Suzuki Y. Pharmacological activation of SERCA ameliorates dystrophic phenotypes in dystrophin-deficient mdx mice. Hum Mol Genet 2021; 30:1006-1019. [PMID: 33822956 PMCID: PMC8170845 DOI: 10.1093/hmg/ddab100] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscular weakness because of the loss of dystrophin. Extracellular Ca2+ flows into the cytoplasm through membrane tears in dystrophin-deficient myofibers, which leads to muscle contracture and necrosis. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) takes up cytosolic Ca2+ into the sarcoplasmic reticulum, but its activity is decreased in dystrophic muscle. Here, we show that an allosteric SERCA activator, CDN1163, ameliorates dystrophic phenotypes in dystrophin-deficient mdx mice. The administration of CDN1163 prevented exercise-induced muscular damage and restored mitochondrial function. In addition, treatment with CDN1163 for 7 weeks enhanced muscular strength and reduced muscular degeneration and fibrosis in mdx mice. Our findings provide preclinical proof-of-concept evidence that pharmacological activation of SERCA could be a promising therapeutic strategy for DMD. Moreover, CDN1163 improved muscular strength surprisingly in wild-type mice, which may pave the new way for the treatment of muscular dysfunction.
Collapse
Affiliation(s)
- Ken'ichiro Nogami
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Maruyama
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Fusako Sakai-Takemura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ahmed Elhussieny
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neurology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Michihiro Imamura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Megumu Ogawa
- Department of Neuromuscular Research, National Institute of Neuroscience, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Clinical Development, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuki Tamura
- Graduate School of Health and Sport Science, Nippon Sport Science University, Tokyo, Japan.,Research Institute for Sport Science, Nippon Sport Science University, Tokyo, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | - Yuko Miyagoe-Suzuki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
7
|
A Phase 1/2 Study of Flavocoxid, an Oral NF-κB Inhibitor, in Duchenne Muscular Dystrophy. Brain Sci 2021; 11:brainsci11010115. [PMID: 33467104 PMCID: PMC7830560 DOI: 10.3390/brainsci11010115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
Flavocoxid is a blended extract containing baicalin and catechin with potent antioxidant and anti-inflammatory properties due to the inhibition of the cyclooxygenase (COX) and 5-lipoxygenase (5-LOX) enzymes, nuclear factor-κB (NF-κB), tumor necrosis factor (TNF)-alpha, and the mitogen-activated protein kinases (MAPKs) pathways. This phase 1/2 study was designed to assess the safety and tolerability of flavocoxid in patients with Duchenne muscular dystrophy (DMD). Thirty-four patients were recruited: 17 were treated with flavocoxid at an oral dose of 250 or 500 mg, according to body weight, for one year; 17 did not receive flavocoxid and served as controls. The treatment was well tolerated and nobody dropped out. Flavocoxid induced a significant reduction in serum interleukin (IL)-1 beta and TNF-alpha only in the group of DMD boys on add-on therapy (flavocoxid added to steroids for at least six months). The decrease in IL-1 beta was higher in younger boys. The serum H2O2 concentrations significantly decreased in patients treated with flavocoxid alone with a secondary reduction of serum glutathione peroxidase (GPx) levels, especially in younger boys. The exploratory outcome measures failed to show significant effects but there was a trend showing that the younger boys who received treatment were faster at performing the Gowers' maneuver, while the older boys who received treatment were faster at doing the 10-m walk test (10MWT). Therefore, a double-blind, placebo-controlled study for at least two/three years is warranted to verify flavocoxid as a steroid substitute or as add-on therapy to steroids.
Collapse
|
8
|
Nagy S, Hafner P, Schmidt S, Rubino-Nacht D, Schädelin S, Bieri O, Fischer D. Tamoxifen in Duchenne muscular dystrophy (TAMDMD): study protocol for a multicenter, randomized, placebo-controlled, double-blind phase 3 trial. Trials 2019; 20:637. [PMID: 31752977 PMCID: PMC6869203 DOI: 10.1186/s13063-019-3740-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/21/2019] [Indexed: 12/25/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an inherited neuromuscular disorder of childhood with a devastating disease course. Several targeted gene therapies and molecular approaches have been or are currently being tested in clinical trials; however, a causative therapy is still not available and best supportive care is limited to oral glucocorticoids with numerous long-term side effects. Tamoxifen is a selective estrogen receptor regulator, and shows antioxidant actions and regulatory roles in the calcium homeostasis besides its antitumor activity. In a mouse model of DMD, oral tamoxifen significantly improved muscle strength and reduced muscle fatigue. This multicenter, randomized, double-blind, placebo-controlled phase III trial aims to demonstrate safety and efficacy of tamoxifen over placebo in pediatric patients with DMD. After completion of the double-blind phase, an open-label extension of the study will be offered to all participants. Methods/design At least 71 ambulant and up to 20 nonambulant patients with DMD are planned to be enrolled at multiple European sites. Patients will be randomly assigned to receive either tamoxifen 20 mg or placebo daily over 48 weeks. In the open-label extension phase, all patients will be offered tamoxifen for a further 48 weeks. The primary endpoint of the double-blind phase is defined as the change of the D1 domain of the motor function measure in ambulant patients or a change of the D2 domain in nonambulant patients under tamoxifen compared to placebo. Secondary outcome measures include change in timed function tests, quantitative muscle testing, and quantitative magnetic resonance imaging of thigh muscles. Laboratory analyses including biomarkers of tamoxifen metabolism and muscle dystrophy will also be assessed. Discussion The aim of the study is to investigate whether tamoxifen can reduce disease progression in ambulant and nonambulant patients with DMD over 48 weeks. Motor function measures comprise the primary endpoint, whereas further clinical and radiological assessments and laboratory biomarkers are performed to provide more data on safety and efficacy. An adjacent open-label extension phase is planned to test if earlier initiation of the treatment with tamoxifen (verum arm of double-blind phase) compared to a delayed start can reduce disease progression more efficiently. Trial registration ClinicalTrials.gov, NCT03354039. Registered on 27 November 2017.
Collapse
Affiliation(s)
- Sara Nagy
- Division of Developmental- and Neuropaediatrics, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031, Basel, Switzerland. .,Department of Neurology, University Hospital Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland.
| | - Patricia Hafner
- Division of Developmental- and Neuropaediatrics, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031, Basel, Switzerland
| | - Simone Schmidt
- Division of Developmental- and Neuropaediatrics, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031, Basel, Switzerland
| | - Daniela Rubino-Nacht
- Division of Developmental- and Neuropaediatrics, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031, Basel, Switzerland
| | - Sabine Schädelin
- Clinical Trial Unit, University of Basel, Schanzenstrasse 55, 4056, Basel, Switzerland
| | - Oliver Bieri
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, University of Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Dirk Fischer
- Division of Developmental- and Neuropaediatrics, University Children's Hospital Basel (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031, Basel, Switzerland
| |
Collapse
|
9
|
Iskenderian A, Liu N, Deng Q, Huang Y, Shen C, Palmieri K, Crooker R, Lundberg D, Kastrapeli N, Pescatore B, Romashko A, Dumas J, Comeau R, Norton A, Pan J, Rong H, Derakhchan K, Ehmann DE. Myostatin and activin blockade by engineered follistatin results in hypertrophy and improves dystrophic pathology in mdx mouse more than myostatin blockade alone. Skelet Muscle 2018; 8:34. [PMID: 30368252 PMCID: PMC6204036 DOI: 10.1186/s13395-018-0180-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 10/14/2018] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Myostatin antagonists are being developed as therapies for Duchenne muscular dystrophy due to their strong hypertrophic effects on skeletal muscle. Engineered follistatin has the potential to combine the hypertrophy of myostatin antagonism with the anti-inflammatory and anti-fibrotic effects of activin A antagonism. METHODS Engineered follistatin was administered to C57BL/6 mice for 4 weeks, and muscle mass and myofiber size was measured. In the mdx model, engineered follistatin was dosed for 12 weeks in two studies comparing to an Fc fusion of the activin IIB receptor or an anti-myostatin antibody. Functional measurements of grip strength and tetanic force were combined with tissue analysis for markers of necrosis, inflammation, and fibrosis to evaluate improvement in dystrophic pathology. RESULTS In wild-type and mdx mice, dose-dependent increases in muscle mass and quadriceps myofiber size were observed for engineered follistatin. In mdx, increases in grip strength and tetanic force were combined with improvements in muscle markers for necrosis, inflammation, and fibrosis. Improvements in dystrophic pathology were greater for engineered follistatin than the anti-myostatin antibody. CONCLUSIONS Engineered follistatin generated hypertrophy and anti-fibrotic effects in the mdx model.
Collapse
Affiliation(s)
- Andrea Iskenderian
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Nan Liu
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Qingwei Deng
- Research, Shire Pharmaceuticals, Lexington, MA, 02421, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Yan Huang
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Chuan Shen
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Kathleen Palmieri
- Research, Shire Pharmaceuticals, Lexington, MA, 02421, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Robert Crooker
- Research, Shire Pharmaceuticals, Lexington, MA, 02421, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Dianna Lundberg
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Niksa Kastrapeli
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Brian Pescatore
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Alla Romashko
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - John Dumas
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Robert Comeau
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Angela Norton
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Jing Pan
- Discovery Therapeutics, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Haojing Rong
- Nonclinical Development, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - Katayoun Derakhchan
- Nonclinical Development, Shire Pharmaceuticals, Lexington, MA, USA.,Drug Discovery, Shire, Cambridge, MA, USA
| | - David E Ehmann
- Research, Shire Pharmaceuticals, Lexington, MA, 02421, USA. .,Drug Discovery, Shire, Cambridge, MA, USA.
| |
Collapse
|
10
|
Ismail HM, Dorchies OM, Scapozza L. The potential and benefits of repurposing existing drugs to treat rare muscular dystrophies. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1452733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hesham M. Ismail
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Olivier M. Dorchies
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| |
Collapse
|
11
|
Barnard AM, Willcocks RJ, Finanger EL, Daniels MJ, Triplett WT, Rooney WD, Lott DJ, Forbes SC, Wang DJ, Senesac CR, Harrington AT, Finkel RS, Russman BS, Byrne BJ, Tennekoon GI, Walter GA, Sweeney HL, Vandenborne K. Skeletal muscle magnetic resonance biomarkers correlate with function and sentinel events in Duchenne muscular dystrophy. PLoS One 2018; 13:e0194283. [PMID: 29554116 PMCID: PMC5858773 DOI: 10.1371/journal.pone.0194283] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To provide evidence for quantitative magnetic resonance (qMR) biomarkers in Duchenne muscular dystrophy by investigating the relationship between qMR measures of lower extremity muscle pathology and functional endpoints in a large ambulatory cohort using a multicenter study design. METHODS MR spectroscopy and quantitative imaging were implemented to measure intramuscular fat fraction and the transverse magnetization relaxation time constant (T2) in lower extremity muscles of 136 participants with Duchenne muscular dystrophy. Measures were collected at 554 visits over 48 months at one of three imaging sites. Fat fraction was measured in the soleus and vastus lateralis using MR spectroscopy, while T2 was assessed using MRI in eight lower extremity muscles. Ambulatory function was measured using the 10m walk/run, climb four stairs, supine to stand, and six minute walk tests. RESULTS Significant correlations were found between all qMR and functional measures. Vastus lateralis qMR measures correlated most strongly to functional endpoints (|ρ| = 0.68-0.78), although measures in other rapidly progressing muscles including the biceps femoris (|ρ| = 0.63-0.73) and peroneals (|ρ| = 0.59-0.72) also showed strong correlations. Quantitative MR biomarkers were excellent indicators of loss of functional ability and correlated with qualitative measures of function. A VL FF of 0.40 was an approximate lower threshold of muscle pathology associated with loss of ambulation. DISCUSSION Lower extremity qMR biomarkers have a robust relationship to clinically meaningful measures of ambulatory function in Duchenne muscular dystrophy. These results provide strong supporting evidence for qMR biomarkers and set the stage for their potential use as surrogate outcomes in clinical trials.
Collapse
Affiliation(s)
- Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Erika L. Finanger
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, FL, United States of America
| | - William T. Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Dah-Jyuu Wang
- Department of Radiology, Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Ann T. Harrington
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | | | - Barry S. Russman
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Barry J. Byrne
- Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, FL, United States of America
| | - Gihan I. Tennekoon
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States of America
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States of America
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
12
|
Bell EL, Shine RW, Dwyer P, Olson L, Truong J, Fredenburg R, Goddeeris M, Stickens D, Tozzo E. PPARδ modulation rescues mitochondrial fatty acid oxidation defects in the mdx model of muscular dystrophy. Mitochondrion 2018; 46:51-58. [PMID: 29458111 DOI: 10.1016/j.mito.2018.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/05/2018] [Accepted: 02/15/2018] [Indexed: 12/25/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a recessive, fatal X-linked disease that is characterized by progressive skeletal muscle wasting due to the absence of dystrophin, which is an a essential protein that bridges the inner cytoskeleton and extra-cellular matrix. This study set out to characterize the mitochondria in primary muscle satellite cell derived myoblasts from mdx mice and wild type control mice. Compared to wild type derived cells the mdx derived cells have reduced mitochondrial bioenergetics and have fewer mitochondria. Here, we demonstrate that a novel PPARδ modulator improves mitochondrial function in the mdx mice, which supports that modulating PPARδ may be therapeutically beneficial in DMD patients.
Collapse
Affiliation(s)
- Eric L Bell
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States.
| | - Robert W Shine
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Peter Dwyer
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Lyndsay Olson
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Jennifer Truong
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Ross Fredenburg
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Matthew Goddeeris
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Dominique Stickens
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| | - Effie Tozzo
- Mitobridge, Inc, 1030 Massachusetts Avenue, Cambridge, MA 02138, United States
| |
Collapse
|
13
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
14
|
Pambianco S, Giovarelli M, Perrotta C, Zecchini S, Cervia D, Di Renzo I, Moscheni C, Ripolone M, Violano R, Moggio M, Bassi MT, Puri PL, Latella L, Clementi E, De Palma C. Reversal of Defective Mitochondrial Biogenesis in Limb-Girdle Muscular Dystrophy 2D by Independent Modulation of Histone and PGC-1α Acetylation. Cell Rep 2017; 17:3010-3023. [PMID: 27974213 DOI: 10.1016/j.celrep.2016.11.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/10/2016] [Accepted: 11/11/2016] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction occurs in many muscle degenerative disorders. Here, we demonstrate that mitochondrial biogenesis was impaired in limb-girdle muscular dystrophy (LGMD) 2D patients and mice and was associated with impaired OxPhos capacity. Two distinct approaches that modulated histones or peroxisome proliferator-activated receptor-gamma coactivator 1 α (PGC-1α) acetylation exerted equivalent functional effects by targeting different mitochondrial pathways (mitochondrial biogenesis or fatty acid oxidation[FAO]). The histone deacetylase inhibitor Trichostatin A (TSA) changed chromatin assembly at the PGC-1α promoter, restored mitochondrial biogenesis, and enhanced muscle oxidative capacity. Conversely, nitric oxide (NO) triggered post translation modifications of PGC-1α and induced FAO, recovering the bioenergetics impairment of muscles but shunting the defective mitochondrial biogenesis. In conclusion, a transcriptional blockade of mitochondrial biogenesis occurred in LGMD-2D and could be recovered by TSA changing chromatin conformation, or it could be overcome by NO activating a mitochondrial salvage pathway.
Collapse
Affiliation(s)
- Sarah Pambianco
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy
| | - Davide Cervia
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy; Department for Innovation in Biological, Agro-food and Forest systems, Università degli Studi della Tuscia, 01100 Viterbo, Italy
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences "Luigi Sacco," Università degli Studi di Milano, 20157 Milano, Italy
| | - Michela Ripolone
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | - Raffaella Violano
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | - Maurizio Moggio
- Neuromuscular Unit, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, 20122 Milano, Italy
| | | | - Pier Lorenzo Puri
- Epigenetics and Regenerative Pharmacology, IRCCS Fondazione Santa Lucia, 00142 Roma, Italy; Sanford Children's Health Research Center, Sanford Prebys Burnham Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Lucia Latella
- Epigenetics and Regenerative Pharmacology, IRCCS Fondazione Santa Lucia, 00142 Roma, Italy; National Research Council-Institute of Translational Pharmacology, 00179 Roma, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy; IRCCS Eugenio Medea, 23842 Bosisio Parini, Italy.
| | - Clara De Palma
- Department of Biomedical and Clinical Sciences, Unit of Clinical Pharmacology, University Hospital "Luigi Sacco"-ASST Fatebenefratelli Sacco, National Research Council-Institute of Neuroscience, Università degli Studi di Milano, 20157 Milano, Italy.
| |
Collapse
|
15
|
Van Ry PM, Fontelonga TM, Barraza-Flores P, Sarathy A, Nunes AM, Burkin DJ. ECM-Related Myopathies and Muscular Dystrophies: Pros and Cons of Protein Therapies. Compr Physiol 2017; 7:1519-1536. [PMID: 28915335 DOI: 10.1002/cphy.c150033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Extracellular matrix (ECM) myopathies and muscular dystrophies are a group of genetic diseases caused by mutations in genes encoding proteins that provide critical links between muscle cells and the extracellular matrix. These include structural proteins of the ECM, muscle cell receptors, enzymes, and intracellular proteins. Loss of adhesion within the myomatrix results in progressive muscle weakness. For many ECM muscular dystrophies, symptoms can occur any time after birth and often result in reduced life expectancy. There are no cures for the ECM-related muscular dystrophies and treatment options are limited to palliative care. Several therapeutic approaches have been explored to treat muscular dystrophies including gene therapy, gene editing, exon skipping, embryonic, and adult stem cell therapy, targeting genetic modifiers, modulating inflammatory responses, or preventing muscle degeneration. Recently, protein therapies that replace components of the defective myomatrix or enhance muscle and/or extracellular matrix integrity and function have been explored. Preclinical studies for many of these biologics have been promising in animal models of these muscle diseases. This review aims to summarize the ECM muscular dystrophies for which protein therapies are being developed and discuss the exciting potential and possible limitations of this approach for treating this family of devastating genetic muscle diseases. © 2017 American Physiological Society. Compr Physiol 7:1519-1536, 2017.
Collapse
Affiliation(s)
- Pam M Van Ry
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Tatiana M Fontelonga
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Pamela Barraza-Flores
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Apurva Sarathy
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Andreia M Nunes
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA.,Departamento de Biologia Animal, Centro de Ecologia, Evolucao e Alteracoes Ambientais, Faculdade de Ciencias, Universidade de Lisboa, Lisbon, Portugal
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| |
Collapse
|
16
|
Affiliation(s)
- Danial E. Baker
- Drug Information Center, and Pharmacy Practice, College of Pharmacy, Washington State University Spokane
| |
Collapse
|
17
|
Reduced serum myostatin concentrations associated with genetic muscle disease progression. J Neurol 2017; 264:541-553. [DOI: 10.1007/s00415-016-8379-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 12/26/2022]
|
18
|
Tsao J, Kovanecz I, Awadalla N, Gelfand R, Sinha-Hikim I, White RA, Gonzalez-Cadavid NF. Muscle Derived Stem Cells Stimulate Muscle Myofiber Repair and Counteract Fat Infiltration in a Diabetic Mouse Model of Critical Limb Ischemia. ACTA ACUST UNITED AC 2016; 6. [PMID: 28217409 PMCID: PMC5313052 DOI: 10.4172/2157-7633.1000370] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Critical Limb Ischemia (CLI) affects patients with Type 2 Diabetes (T2D) and obesity, with high risk of amputation and post-surgical mortality, and no effective medical treatment. Stem cell therapy, mainly with bone marrow mesenchymal, adipose derived, endothelial, hematopoietic, and umbilical cord stem cells, is promising in CLI mouse and rat models and is in clinical trials. Their general focus is on angiogenic repair, with no reports on the alleviation of necrosis, lipofibrosis, and myofiber regeneration in the ischemic muscle, or the use of Muscle Derived Stem Cells (MDSC) alone or in combination with pharmacological adjuvants, in the context of CLI in T2D. Methods Using a T2D mouse model of CLI induced by severe unilateral femoral artery ligation, we tested: a) the repair efficacy of MDSC implanted into the ischemic muscle and the effects of concurrent intraperitoneal administration of a nitric oxide generator, molsidomine; and b) whether MDSC may partially counteract their own repair effects by stimulating the expression of myostatin, the main lipofibrotic agent in the muscle and inhibitor of muscle mass. Results MDSC: a) reduced mortality, and b) in the ischemic muscle, increased stem cell number and myofiber central nuclei, reduced fat infiltration, myofibroblast number, and myofiber apoptosis, and increased smooth muscle and endothelial cells, as well as neurotrophic factors. The content of myosin heavy chain 2 (MHC-2) myofibers was not restored and collagen was increased, in association with myostatin overexpression. Supplementation of MDSC with molsidomine failed to stimulate the beneficial effects of MDSC, except for some reduction in myostatin overexpression. Molsidomine given alone was rather ineffective, except for inhibiting apoptosis and myostatin overexpression. Conclusions MDSC improved CLI muscle repair, but molsidomine did not stimulate this process. The combination of MDSC with anti-myostatin approaches should be explored to restore myofiber MHC composition.
Collapse
Affiliation(s)
- J Tsao
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - I Kovanecz
- Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - N Awadalla
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - R Gelfand
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA; Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - I Sinha-Hikim
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
| | - R A White
- Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - N F Gonzalez-Cadavid
- Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA; Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
19
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
20
|
Miyatake S, Shimizu-Motohashi Y, Takeda S, Aoki Y. Anti-inflammatory drugs for Duchenne muscular dystrophy: focus on skeletal muscle-releasing factors. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2745-58. [PMID: 27621596 PMCID: PMC5012616 DOI: 10.2147/dddt.s110163] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD), an incurable and a progressive muscle wasting disease, is caused by the absence of dystrophin protein, leading to recurrent muscle fiber damage during contraction. The inflammatory response to fiber damage is a compelling candidate mechanism for disease exacerbation. The only established pharmacological treatment for DMD is corticosteroids to suppress muscle inflammation, however this treatment is limited by its insufficient therapeutic efficacy and considerable side effects. Recent reports show the therapeutic potential of inhibiting or enhancing pro- or anti-inflammatory factors released from DMD skeletal muscles, resulting in significant recovery from muscle atrophy and dysfunction. We discuss and review the recent findings of DMD inflammation and opportunities for drug development targeting specific releasing factors from skeletal muscles. It has been speculated that nonsteroidal anti-inflammatory drugs targeting specific inflammatory factors are more effective and have less side effects for DMD compared with steroidal drugs. For example, calcium channels, reactive oxygen species, and nuclear factor-κB signaling factors are the most promising targets as master regulators of inflammatory response in DMD skeletal muscles. If they are combined with an oligonucleotide-based exon skipping therapy to restore dystrophin expression, the anti-inflammatory drug therapies may address the present therapeutic limitation of low efficiency for DMD.
Collapse
Affiliation(s)
- Shouta Miyatake
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuko Shimizu-Motohashi
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
21
|
Activation of Wnt3a signaling promotes myogenic differentiation of mesenchymal stem cells in mdx mice. Acta Pharmacol Sin 2016; 37:873-81. [PMID: 27133298 DOI: 10.1038/aps.2016.38] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/04/2016] [Indexed: 02/08/2023]
Abstract
AIM Duchenne muscular dystrophy (DMD) is an X-linked genetic muscular disorder with no effective treatment at present. Mesenchymal stem cell (MSC) transplantation has been used to treat DMD, but the efficiency is low. Our previous studies show that activation of Wnt3a signaling promotes myogenic differentiation of MSCs in vitro. Here we report an effective MSC transplantation therapy in mdx mice by activation of Wnt3a signaling. METHODS MSCs were isolated from mouse bone marrow, and pretreated with Wnt3a-conditioned medium (Wnt3a-CM), then transplanted into mdx mice. The recipient mice were euthanized at 4, 8, 12, 16 weeks after the transplantation, and muscle pathological changes were examined. The expression of dystrophin in muscle was detected using immunofluorescence staining, RT-PCR and Western blotting. RESULTS Sixteen weeks later, transplantation of Wnt3a-pretreated MSCs in mdx mice improved the characteristics of dystrophic muscles evidenced by significant reductions in centrally nucleated myofibers, the variability range of cross-sectional area (CSA) and the connective tissue area of myofibers. Furthermore, transplantation of Wnt3a-pretreated MSCs in mdx mice gradually and markedly increased the expression of dystrophin in muscle, and improved the efficiency of myogenic differentiation. CONCLUSION Transplantation of Wnt3a-pretreated MSCs in mdx mice results in long-term amelioration of the dystrophic phenotype and restores dystrophin expression in muscle. The results suggest that Wnt3a may be a promising candidate for the treatment of DMD.
Collapse
|
22
|
Cossu G, Previtali SC, Napolitano S, Cicalese MP, Tedesco FS, Nicastro F, Noviello M, Roostalu U, Natali Sora MG, Scarlato M, De Pellegrin M, Godi C, Giuliani S, Ciotti F, Tonlorenzi R, Lorenzetti I, Rivellini C, Benedetti S, Gatti R, Marktel S, Mazzi B, Tettamanti A, Ragazzi M, Imro MA, Marano G, Ambrosi A, Fiori R, Sormani MP, Bonini C, Venturini M, Politi LS, Torrente Y, Ciceri F. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol Med 2016; 7:1513-28. [PMID: 26543057 PMCID: PMC4693504 DOI: 10.15252/emmm.201505636] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intra‐arterial transplantation of mesoangioblasts proved safe and partially efficacious in preclinical models of muscular dystrophy. We now report the first‐in‐human, exploratory, non‐randomized open‐label phase I–IIa clinical trial of intra‐arterial HLA‐matched donor cell transplantation in 5 Duchenne patients. We administered escalating doses of donor‐derived mesoangioblasts in limb arteries under immunosuppressive therapy (tacrolimus). Four consecutive infusions were performed at 2‐month intervals, preceded and followed by clinical, laboratory, and muscular MRI analyses. Two months after the last infusion, a muscle biopsy was performed. Safety was the primary endpoint. The study was relatively safe: One patient developed a thalamic stroke with no clinical consequences and whose correlation with mesoangioblast infusion remained unclear. MRI documented the progression of the disease in 4/5 patients. Functional measures were transiently stabilized in 2/3 ambulant patients, but no functional improvements were observed. Low level of donor DNA was detected in muscle biopsies of 4/5 patients and donor‐derived dystrophin in 1. Intra‐arterial transplantation of donor mesoangioblasts in human proved to be feasible and relatively safe. Future implementation of the protocol, together with a younger age of patients, will be needed to approach efficacy.
Collapse
Affiliation(s)
- Giulio Cossu
- Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Stefano C Previtali
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Napolitano
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Cicalese
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesca Nicastro
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maddalena Noviello
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Urmas Roostalu
- Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | | | - Marina Scarlato
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Claudia Godi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy Neuroradiology Department and Neuroradiology Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Giuliani
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ciotti
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rossana Tonlorenzi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Isabella Lorenzetti
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Rivellini
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Benedetti
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Roberto Gatti
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Benedetta Mazzi
- Immunogenetics Laboratory, Department of Immunohematology & Blood Transfusion, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Tettamanti
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, London, UK
| | | | | | | | - Rossana Fiori
- Unit of Anesthesiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Venturini
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Letterio S Politi
- Neuroradiology Department and Neuroradiology Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Ciceri
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Engaging Participants in Rare Disease Research: A Qualitative Study of Duchenne Muscular Dystrophy. Clin Ther 2016; 38:1474-1484.e2. [DOI: 10.1016/j.clinthera.2016.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023]
|
24
|
Adolescent idiopathic scoliosis: evidence for intrinsic factors driving aetiology and progression. INTERNATIONAL ORTHOPAEDICS 2016; 40:2075-2080. [PMID: 26961194 DOI: 10.1007/s00264-016-3132-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/08/2016] [Indexed: 12/22/2022]
Abstract
Adolescent idiopathic scoliosis (AIS) is now considered to be a multifactorial heterogeneous disease, with recent genomic studies supporting the role of intrinsic factors in contributing to the onset of disease pathology and curve progression. Understanding the key molecular signalling pathways by which these intrinsic factors mediate AIS pathology may facilitate the development of pharmacological therapeutics and the identification of predictive markers of progression. The heterogenic nature of AIS has implicated multiple tissue types in the disease pathophysiology, including spinal bone, intervertebral disc and paraspinal muscles. In this review, we highlight some of the mechanisms and intrinsic molecular regulators within these different tissue types and review the evidence for their involvement in AIS pathology.
Collapse
|
25
|
Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiol Rev 2016; 96:253-305. [PMID: 26676145 DOI: 10.1152/physrev.00007.2015] [Citation(s) in RCA: 309] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Dystrophin is a long rod-shaped protein that connects the subsarcolemmal cytoskeleton to a complex of proteins in the surface membrane (dystrophin protein complex, DPC), with further connections via laminin to other extracellular matrix proteins. Initially considered a structural complex that protected the sarcolemma from mechanical damage, the DPC is now known to serve as a scaffold for numerous signaling proteins. Absence or reduced expression of dystrophin or many of the DPC components cause the muscular dystrophies, a group of inherited diseases in which repeated bouts of muscle damage lead to atrophy and fibrosis, and eventually muscle degeneration. The normal function of dystrophin is poorly defined. In its absence a complex series of changes occur with multiple muscle proteins showing reduced or increased expression or being modified in various ways. In this review, we will consider the various proteins whose expression and function is changed in muscular dystrophies, focusing on Ca(2+)-permeable channels, nitric oxide synthase, NADPH oxidase, and caveolins. Excessive Ca(2+) entry, increased membrane permeability, disordered caveolar function, and increased levels of reactive oxygen species are early changes in the disease, and the hypotheses for these phenomena will be critically considered. The aim of the review is to define the early damage pathways in muscular dystrophy which might be appropriate targets for therapy designed to minimize the muscle degeneration and slow the progression of the disease.
Collapse
Affiliation(s)
- David G Allen
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Nicholas P Whitehead
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| | - Stanley C Froehner
- Sydney Medical School & Bosch Institute, University of Sydney, New South Wales, Australia; and Department of Physiology & Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
26
|
Kosir AM, Mader TL, Greising AG, Novotny SA, Baltgalvis KA, Lowe DA. Influence of ovarian hormones on strength loss in healthy and dystrophic female mice. Med Sci Sports Exerc 2016; 47:1177-87. [PMID: 25255128 DOI: 10.1249/mss.0000000000000531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE The primary objective of this study was to determine whether strength loss and recovery after eccentric contractions are impaired in healthy and dystrophic female mice with low levels of ovarian hormones. METHODS Female C57BL/6 (wild-type) or mdx mice were randomly assigned to ovarian-intact (Sham) and ovariectomized (Ovx) groups. Anterior crural muscles were tested for susceptibility to injury from 150 or 50 eccentric contractions in wild-type and mdx mice, respectively. An additional experiment challenged mdx mice with a 2-wk treadmill running protocol followed by an eccentric contraction injury to posterior crural muscles. Functional recovery from injury was evaluated in wild-type mice by measuring isometric torque 3, 7, 14, or 21 d after injury. RESULTS Ovarian hormone deficiency in wild-type mice did not affect susceptibility to injury because the ∼50% isometric torque loss after eccentric contractions did not differ between Sham and Ovx mice (P = 0.121). Similarly, in mdx mice, hormone deficiency did not affect the percent of preinjury isometric torque lost by anterior crural muscles after eccentric contractions (P = 0.952), but the percent of preinjury torque in posterior crural muscles was lower in Ovx than in Sham mice (P = 0.014). Recovery from injury in wild-type mice was affected by hormone deficiency. Sham mice recovered preinjury isometric strength by 14 d (96% ± 2%), whereas Ovx mice maintained deficits at 14 and 21 d after injury (80% ± 3% and 84% ± 2%, P < 0.001). CONCLUSIONS Ovarian hormone status did not affect the vulnerability of skeletal muscle to strength loss after eccentric contractions. However, ovarian hormone deficiency did impair the recovery of muscle strength in female mice.
Collapse
Affiliation(s)
- Allison M Kosir
- Programs in Physical Therapy and Rehabilitation Science, Department of Physical Medicine and Rehabilitation, Medical School, University of Minnesota, Minneapolis, MN
| | | | | | | | | | | |
Collapse
|
27
|
Briata P, Bordo D, Puppo M, Gorlero F, Rossi M, Perrone-Bizzozero N, Gherzi R. Diverse roles of the nucleic acid-binding protein KHSRP in cell differentiation and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 7:227-40. [PMID: 26708421 DOI: 10.1002/wrna.1327] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 12/15/2022]
Abstract
The single-stranded nucleic acid-binding protein KHSRP (KH-type splicing regulatory protein) modulates RNA life and gene expression at various levels. KHSRP controls important cellular functions as different as proliferation, differentiation, metabolism, and response to infectious agents. We summarize and discuss experimental evidence providing a potential link between changes in KHSRP expression/function and human diseases including neuromuscular disorders, obesity, type II diabetes, and cancer.
Collapse
Affiliation(s)
- Paola Briata
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Domenico Bordo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Margherita Puppo
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Franco Gorlero
- S.C. Ginecologia e Ostetricia Galliera Hospital, Genova, Italy.,School of Medicine, DINOGMI, University of Genova, Genova, Italy
| | - Martina Rossi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| | - Nora Perrone-Bizzozero
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Roberto Gherzi
- Gene Expression Regulation Laboratory, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|
28
|
Abstract
The human transcriptome is composed of a vast RNA population that undergoes further diversification by splicing. Detecting specific splice sites in this large sequence pool is the responsibility of the major and minor spliceosomes in collaboration with numerous splicing factors. This complexity makes splicing susceptible to sequence polymorphisms and deleterious mutations. Indeed, RNA mis-splicing underlies a growing number of human diseases with substantial societal consequences. Here, we provide an overview of RNA splicing mechanisms followed by a discussion of disease-associated errors, with an emphasis on recently described mutations that have provided new insights into splicing regulation. We also discuss emerging strategies for splicing-modulating therapy.
Collapse
Affiliation(s)
- Marina M Scotti
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida 32610-3610 USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida 32610-3610 USA
| |
Collapse
|
29
|
Hyzewicz J, Ruegg UT, Takeda S. Comparison of Experimental Protocols of Physical Exercise for mdx Mice and Duchenne Muscular Dystrophy Patients. J Neuromuscul Dis 2015; 2:325-342. [PMID: 27858750 PMCID: PMC5240598 DOI: 10.3233/jnd-150106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is caused by mutations in the gene coding for dystrophin and leads to muscle degeneration, wheelchair dependence and death by cardiac or respiratory failure. Physical exercise has been proposed as a palliative therapy for DMD to maintain muscle strength and prevent contractures for as long as possible. However, its practice remains controversial because the benefits of training may be counteracted by muscle overuse and damage. The effects of physical exercise have been investigated in muscles of dystrophin-deficient mdx mice and in patients with DMD. However, a lack of uniformity among protocols limits comparability between studies and translatability of results from animals to humans. In the present review, we summarize and discuss published protocols used to investigate the effects of physical exercise on mdx mice and DMD patients, with the objectives of improving comparability between studies and identifying future research directions.
Collapse
Affiliation(s)
- Janek Hyzewicz
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
30
|
Massouridès E, Polentes J, Mangeot PE, Mournetas V, Nectoux J, Deburgrave N, Nusbaum P, Leturcq F, Popplewell L, Dickson G, Wein N, Flanigan KM, Peschanski M, Chelly J, Pinset C. Dp412e: a novel human embryonic dystrophin isoform induced by BMP4 in early differentiated cells. Skelet Muscle 2015; 5:40. [PMID: 26568816 PMCID: PMC4644319 DOI: 10.1186/s13395-015-0062-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/21/2015] [Indexed: 01/09/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a devastating X-linked recessive genetic myopathy. DMD physiopathology is still not fully understood and a prenatal onset is suspected but difficult to address. Methods The bone morphogenetic protein 4 (BMP4) is a critical signaling molecule involved in mesoderm commitment. Human induced pluripotent stem cells (hiPSCs) from DMD and healthy individuals and human embryonic stem cells (hESCs) treated with BMP4 allowed us to model the early steps of myogenesis in normal and DMD contexts. Results Unexpectedly, 72h following BMP4 treatment, a new long DMD transcript was detected in all tested hiPSCs and hESCs, at levels similar to that found in adult skeletal muscle. This novel transcript named “Dp412e” has a specific untranslated first exon which is conserved only in a sub-group of anthropoids including human. The corresponding novel dystrophin protein of 412-kiloDalton (kDa), characterized by an N-terminal-truncated actin-binding domain, was detected in normal BMP4-treated hiPSCs/hESCs and in embryoid bodies. Finally, using a phosphorodiamidate morpholino oligomer (PMO) targeting the DMD exon 53, we demonstrated the feasibility of exon skipping validation with this BMP4-inducible hiPSCs model. Conclusions In this study, the use of hiPSCs to analyze early phases of human development in normal and DMD contexts has led to the discovery of an embryonic 412 kDa dystrophin isoform. Deciphering the regulation process(es) and the function(s) associated to this new isoform can contribute to a better understanding of the DMD physiopathology and potential developmental defects. Moreover, the simple and robust BMP4-inducible model highlighted here, providing large amount of a long DMD transcript and the corresponding protein in only 3 days, is already well-adapted to high-throughput and high-content screening approaches. Therefore, availability of this powerful cell platform can accelerate the development, validation and improvement of DMD genetic therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0062-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jérôme Polentes
- I-STEM, CECS, Génopôle-Campus 1, 5 rue Henri Desbruères, 91030 Evry, Cedex France
| | - Philippe-Emmanuel Mangeot
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France ; Inserm, U1111, Lyon, France ; CNRS, UMR5308, Lyon, France ; Ecole Normale Supérieure de Lyon, Lyon, France ; Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
| | | | - Juliette Nectoux
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Nathalie Deburgrave
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Patrick Nusbaum
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - France Leturcq
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Linda Popplewell
- School of Biological Sciences, Royal Holloway-University of London, Surrey, TW20 0EX UK
| | - George Dickson
- School of Biological Sciences, Royal Holloway-University of London, Surrey, TW20 0EX UK
| | - Nicolas Wein
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205 USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205 USA
| | - Marc Peschanski
- UEVE U861, 91030 Evry, France ; Inserm U861, 91030 Evry, France
| | - Jamel Chelly
- IGBMC-CNRS UMR7104/Inserm U964, 67404 Illkirch, Cedex France
| | | |
Collapse
|
31
|
Sun C, Li S, Li D. Sulforaphane mitigates muscle fibrosis in mdx mice via Nrf2-mediated inhibition of TGF-β/Smad signaling. J Appl Physiol (1985) 2015; 120:377-90. [PMID: 26494449 DOI: 10.1152/japplphysiol.00721.2015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 10/17/2015] [Indexed: 02/06/2023] Open
Abstract
Sulforaphane (SFN), an activator of NF-E2-related factor 2 (Nrf2), has been found to have an antifibrotic effect on liver and lung. However, its effects on dystrophic muscle fibrosis remain unknown. This work was undertaken to evaluate the effects of SFN-mediated activation of Nrf2 on dystrophic muscle fibrosis. Male mdx mice (age 3 mo) were treated with SFN by gavage (2 mg/kg body wt per day) for 3 mo. Experimental results demonstrated that SFN remarkably attenuated skeletal and cardiac muscle fibrosis as indicated by reduced Sirius Red staining and immunostaining of the extracellular matrix. Moreover, SFN significantly inhibited the transforming growth factor-β (TGF-β)/Smad signaling pathway and suppressed profibrogenic gene and protein expressions such as those of α-smooth muscle actin (α-SMA), fibronectin, collagen I, plasminogen activator inhibitor-1 (PAI-1), and tissue inhibitor metalloproteinase-1 (TIMP-1) in an Nrf2-dependent manner. Furthermore, SFN significantly decreased the expression of inflammatory cytokines CD45, TNF-α, and IL-6 in mdx mice. In conclusion, these results show that SFN can attenuate dystrophic muscle fibrosis by Nrf2-mediated inhibition of the TGF-β/Smad signaling pathway, which indicates that Nrf2 may represent a new target for dystrophic muscle fibrosis.
Collapse
Affiliation(s)
- Chengcao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, People's Republic of China; Institute of Global Health, Wuhan University, Wuhan, People's Republic of China; and
| | - Shujun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, People's Republic of China; Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, Wuhan, People's Republic of China
| | - Dejia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
32
|
Genetic mutations strengthen functional association of LAP1 with DYT1 dystonia and muscular dystrophy. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 766:42-7. [DOI: 10.1016/j.mrrev.2015.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 12/30/2022]
|
33
|
|
34
|
Galectin-1 Protein Therapy Prevents Pathology and Improves Muscle Function in the mdx Mouse Model of Duchenne Muscular Dystrophy. Mol Ther 2015; 23:1285-1297. [PMID: 26050991 DOI: 10.1038/mt.2015.105] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/27/2015] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disease caused by mutations in the dystrophin gene, leading to the loss of a critical component of the sarcolemmal dystrophin glycoprotein complex. Galectin-1 is a small 14 kDa protein normally found in skeletal muscle and has been shown to be a modifier of immune response, muscle repair, and apoptosis. Galectin-1 levels are elevated in the muscle of mouse and dog models of DMD. Together, these findings led us to hypothesize that Galectin-1 may serve as a modifier of disease progression in DMD. To test this hypothesis, recombinant mouse Galectin-1 was produced and used to treat myogenic cells and the mdx mouse model of DMD. Here we show that intramuscular and intraperitoneal injections of Galectin-1 into mdx mice prevented pathology and improved muscle function in skeletal muscle. These improvements were a result of enhanced sarcolemmal stability mediated by elevated utrophin and α7β1 integrin protein levels. Together our results demonstrate for the first time that Galectin-1 may serve as an exciting new protein therapeutic for the treatment of DMD.
Collapse
|
35
|
Sciorati C, Clementi E, Manfredi AA, Rovere-Querini P. Fat deposition and accumulation in the damaged and inflamed skeletal muscle: cellular and molecular players. Cell Mol Life Sci 2015; 72:2135-56. [PMID: 25854633 PMCID: PMC11113943 DOI: 10.1007/s00018-015-1857-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 12/16/2022]
Abstract
The skeletal muscle has the capacity to repair damage by the activation and differentiation of fiber sub-laminar satellite cells. Regeneration impairment due to reduced satellite cells number and/or functional capacity leads to fiber substitution with ectopic tissues including fat and fibrous tissue and to the loss of muscle functions. Muscle mesenchymal cells that in physiological conditions sustain or directly contribute to regeneration differentiate in adipocytes in patients with persistent damage and inflammation of the skeletal muscle. These cells comprise the fibro-adipogenic precursors, the PW1-expressing cells and some interstitial cells associated with vessels (pericytes, mesoangioblasts and myoendothelial cells). Resident fibroblasts that are responsible for collagen deposition and extracellular matrix remodeling during regeneration yield fibrotic tissue and can differentiate into adipose cells. Some authors have also proposed that satellite cells themselves could transdifferentiate into adipocytes, although recent results by lineage tracing techniques seem to put this theory to discussion. This review summarizes findings about muscle resident mesenchymal cell differentiation in adipocytes and recapitulates the molecular mediators involved in intramuscular adipose tissue deposition.
Collapse
Affiliation(s)
- Clara Sciorati
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, via Olgettina 58, 20132, Milan, Italy,
| | | | | | | |
Collapse
|
36
|
Kornegay JN, Spurney CF, Nghiem PP, Brinkmeyer-Langford CL, Hoffman EP, Nagaraju K. Pharmacologic management of Duchenne muscular dystrophy: target identification and preclinical trials. ILAR J 2015; 55:119-49. [PMID: 24936034 DOI: 10.1093/ilar/ilu011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked human disorder in which absence of the protein dystrophin causes degeneration of skeletal and cardiac muscle. For the sake of treatment development, over and above definitive genetic and cell-based therapies, there is considerable interest in drugs that target downstream disease mechanisms. Drug candidates have typically been chosen based on the nature of pathologic lesions and presumed underlying mechanisms and then tested in animal models. Mammalian dystrophinopathies have been characterized in mice (mdx mouse) and dogs (golden retriever muscular dystrophy [GRMD]). Despite promising results in the mdx mouse, some therapies have not shown efficacy in DMD. Although the GRMD model offers a higher hurdle for translation, dogs have primarily been used to test genetic and cellular therapies where there is greater risk. Failed translation of animal studies to DMD raises questions about the propriety of methods and models used to identify drug targets and test efficacy of pharmacologic intervention. The mdx mouse and GRMD dog are genetically homologous to DMD but not necessarily analogous. Subcellular species differences are undoubtedly magnified at the whole-body level in clinical trials. This problem is compounded by disparate cultures in clinical trials and preclinical studies, pointing to a need for greater rigor and transparency in animal experiments. Molecular assays such as mRNA arrays and genome-wide association studies allow identification of genetic drug targets more closely tied to disease pathogenesis. Genes in which polymorphisms have been directly linked to DMD disease progression, as with osteopontin, are particularly attractive targets.
Collapse
|
37
|
Fnip1 regulates skeletal muscle fiber type specification, fatigue resistance, and susceptibility to muscular dystrophy. Proc Natl Acad Sci U S A 2014; 112:424-9. [PMID: 25548157 DOI: 10.1073/pnas.1413021112] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mammalian skeletal muscle is broadly characterized by the presence of two distinct categories of muscle fibers called type I "red" slow twitch and type II "white" fast twitch, which display marked differences in contraction strength, metabolic strategies, and susceptibility to fatigue. The relative representation of each fiber type can have major influences on susceptibility to obesity, diabetes, and muscular dystrophies. However, the molecular factors controlling fiber type specification remain incompletely defined. In this study, we describe the control of fiber type specification and susceptibility to metabolic disease by folliculin interacting protein-1 (Fnip1). Using Fnip1 null mice, we found that loss of Fnip1 increased the representation of type I fibers characterized by increased myoglobin, slow twitch markers [myosin heavy chain 7 (MyH7), succinate dehydrogenase, troponin I 1, troponin C1, troponin T1], capillary density, and mitochondria number. Cultured Fnip1-null muscle fibers had higher oxidative capacity, and isolated Fnip1-null skeletal muscles were more resistant to postcontraction fatigue relative to WT skeletal muscles. Biochemical analyses revealed increased activation of the metabolic sensor AMP kinase (AMPK), and increased expression of the AMPK-target and transcriptional coactivator PGC1α in Fnip1 null skeletal muscle. Genetic disruption of PGC1α rescued normal levels of type I fiber markers MyH7 and myoglobin in Fnip1-null mice. Remarkably, loss of Fnip1 profoundly mitigated muscle damage in a murine model of Duchenne muscular dystrophy. These results indicate that Fnip1 controls skeletal muscle fiber type specification and warrant further study to determine whether inhibition of Fnip1 has therapeutic potential in muscular dystrophy diseases.
Collapse
|
38
|
Gintjee TJJ, Magh ASH, Bertoni C. High throughput screening in duchenne muscular dystrophy: from drug discovery to functional genomics. BIOLOGY 2014; 3:752-80. [PMID: 25405319 PMCID: PMC4280510 DOI: 10.3390/biology3040752] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 01/16/2023]
Abstract
Centers for the screening of biologically active compounds and genomic libraries are becoming common in the academic setting and have enabled researchers devoted to developing strategies for the treatment of diseases or interested in studying a biological phenomenon to have unprecedented access to libraries that, until few years ago, were accessible only by pharmaceutical companies. As a result, new drugs and genetic targets have now been identified for the treatment of Duchenne muscular dystrophy (DMD), the most prominent of the neuromuscular disorders affecting children. Although the work is still at an early stage, the results obtained to date are encouraging and demonstrate the importance that these centers may have in advancing therapeutic strategies for DMD as well as other diseases. This review will provide a summary of the status and progress made toward the development of a cure for this disorder and implementing high-throughput screening (HTS) technologies as the main source of discovery. As more academic institutions are gaining access to HTS as a valuable discovery tool, the identification of new biologically active molecules is likely to grow larger. In addition, the presence in the academic setting of experts in different aspects of the disease will offer the opportunity to develop novel assays capable of identifying new targets to be pursued as potential therapeutic options. These assays will represent an excellent source to be used by pharmaceutical companies for the screening of larger libraries providing the opportunity to establish strong collaborations between the private and academic sectors and maximizing the chances of bringing into the clinic new drugs for the treatment of DMD.
Collapse
Affiliation(s)
- Thomas J J Gintjee
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Alvin S H Magh
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Carmen Bertoni
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| |
Collapse
|
39
|
Kornegay JN, Peterson JM, Bogan DJ, Kline W, Bogan JR, Dow JL, Fan Z, Wang J, Ahn M, Zhu H, Styner M, Guttridge DC. NBD delivery improves the disease phenotype of the golden retriever model of Duchenne muscular dystrophy. Skelet Muscle 2014; 4:18. [PMID: 25789154 PMCID: PMC4364341 DOI: 10.1186/2044-5040-4-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/25/2014] [Indexed: 01/19/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene and afflicts skeletal and cardiac muscles. Previous studies showed that DMD is associated with constitutive activation of NF-κB, and in dystrophin-deficient mdx and utrophin/dystrophin (utrn-/-;mdx) double knock out (dko) mouse models, inhibition of NF-κB with the Nemo Binding Domain (NBD) peptide led to significant improvements in both diaphragm and cardiac muscle function. Methods A trial in golden retriever muscular dystrophy (GRMD) canine model of DMD was initiated with four primary outcomes: skeletal muscle function, MRI of pelvic limb muscles, histopathologic features of skeletal muscles, and safety. GRMD and wild type dogs at 2 months of age were treated for 4 months with NBD by intravenous infusions. Results were compared with those collected from untreated GRMD and wild type dogs through a separate, natural history study. Results Results showed that intravenous delivery of NBD in GRMD dogs led to a recovery of pelvic limb muscle force and improvement of histopathologic lesions. In addition, NBD-treated GRMD dogs had normalized postural changes and a trend towards lower tissue injury on magnetic resonance imaging. Despite this phenotypic improvement, NBD administration over time led to infusion reactions and an immune response in both treated GRMD and wild type dogs. Conclusions This GRMD trial was beneficial both in providing evidence that NBD is efficacious in a large animal DMD model and in identifying potential safety concerns that will be informative moving forward with human trials.
Collapse
Affiliation(s)
- Joe N Kornegay
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; Department of Neurology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; The Gene Therapy Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, Mail Stop 4458, College Station, TX, USA
| | - Jennifer M Peterson
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J Bogan
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; The Gene Therapy Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William Kline
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Janet R Bogan
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; The Gene Therapy Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jennifer L Dow
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; The Gene Therapy Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zheng Fan
- Department of Neurology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiahui Wang
- Department of Psychiatry, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mihye Ahn
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Martin Styner
- Department of Psychiatry, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; Department of Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA ; The Ohio State University College of Medicine, 460W. 12th Avenue, Columbus, OH 43210, USA
| |
Collapse
|
40
|
Pillers DAM. A new day for Duchenne's?: The time has come for newborn screening. Mol Genet Metab 2014; 113:11-3. [PMID: 24974086 DOI: 10.1016/j.ymgme.2014.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 06/07/2014] [Indexed: 01/16/2023]
Affiliation(s)
- De-Ann M Pillers
- Department of Pediatrics, University of Wisconsin, Madison, USA.
| |
Collapse
|
41
|
Validation of ambiguous MLPA results by targeted next-generation sequencing discloses a nonsense mutation in the DMD gene. Clin Chim Acta 2014; 436:155-9. [PMID: 24892813 DOI: 10.1016/j.cca.2014.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 01/16/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is the most common inherited muscular disease and caused by mutations in the DMD gene on the X-chromosome. Multiplex ligation-dependent probe amplification (MLPA) is recognized as a convenient and reliable technique to detect exon deletion/duplication mutations in the DMD gene. Here, we applied targeted semi-conductor next-generation sequencing to clarify the cause of ambiguous MLPA results. METHODS Targeted semi-conductor next-generation sequencing was carried out using the Inherited Disease Panel (IDP) on the Ion Torrent Personal Genome Machine (PGM). RESULTS MLPA analysis disclosed unclassifiable relative peak ratio of exon 18 in a DMD boy. His female cousin was indicated to have exon 18 deletion in one allele. To validate these incompatible results, targeted next-generation sequencing was conducted. A nucleotide change, C.2227 C>T creating a premature stop codon, was in exon 18. Concomitantly, both C and T nucleotides were identified in his cousin's genome. Ambiguous values of the relative peak ratio in MLPA were considered due to the one nucleotide mismatch between the genomic sequence and the probe used in MLPA. CONCLUSION Analysis using IDP on PGM disclosed a nonsense mutation in the DMD gene as a cause of ambiguous results of MLPA.
Collapse
|
42
|
Kharraz Y, Guerra J, Pessina P, Serrano AL, Muñoz-Cánoves P. Understanding the process of fibrosis in Duchenne muscular dystrophy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:965631. [PMID: 24877152 PMCID: PMC4024417 DOI: 10.1155/2014/965631] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/08/2014] [Indexed: 02/06/2023]
Abstract
Fibrosis is the aberrant deposition of extracellular matrix (ECM) components during tissue healing leading to loss of its architecture and function. Fibrotic diseases are often associated with chronic pathologies and occur in a large variety of vital organs and tissues, including skeletal muscle. In human muscle, fibrosis is most readily associated with the severe muscle wasting disorder Duchenne muscular dystrophy (DMD), caused by loss of dystrophin gene function. In DMD, skeletal muscle degenerates and is infiltrated by inflammatory cells and the functions of the muscle stem cells (satellite cells) become impeded and fibrogenic cells hyperproliferate and are overactivated, leading to the substitution of skeletal muscle with nonfunctional fibrotic tissue. Here, we review new developments in our understanding of the mechanisms leading to fibrosis in DMD and several recent advances towards reverting it, as potential treatments to attenuate disease progression.
Collapse
Affiliation(s)
- Yacine Kharraz
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Joana Guerra
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Patrizia Pessina
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Antonio L. Serrano
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative diseases (CIBERNED), Institució Catalana de Recerca i Estudis Avançats (ICREA), Doctor Aiguader 83, 08003 Barcelona, Spain
| |
Collapse
|
43
|
Cossu MV, Cattaneo D, Fucile S, Pellegrino P, Baldelli S, Cozzi V, Capetti A, Clementi E. Combined isosorbide dinitrate and ibuprofen as a novel therapy for muscular dystrophies: evidence from Phase I studies in healthy volunteers. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:411-9. [PMID: 24851040 PMCID: PMC4018313 DOI: 10.2147/dddt.s58803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We designed two Phase I studies that assessed healthy volunteers in order to evaluate the safety and to optimize the dosing of the combination of the drugs isosorbide dinitrate, a nitric oxide donor, and ibuprofen, a nonsteroidal antiinflammatory drug. We designed these studies with the aim of designing a Phase II trial to evaluate the drugs’ efficacy in patients affected by Duchenne muscular dystrophy. For the first trial, ISOFEN1, a single-dose, randomized-sequence, open-label, active control, three-treatment cross-over study, was aimed at comparing the pharmacokinetics of ibuprofen 200 mg and isosorbide dinitrate 20 mg when given alone and concomitantly. The pharmacokinetics of ibuprofen given alone versus ibuprofen given concomitantly with isosorbide dinitrate were similar, as documented by the lack of statistically significant differences in the main drug’s pharmacokinetic parameters (time to maximal concentration [Tmax], maximal concentration [Cmax], area under the curve [AUC]0–t, and AUC0–∞). Similarly, we found that the coadministration of ibuprofen did not significantly affect the pharmacokinetics of isosorbide dinitrate. No issues of safety were detected. The second trial, ISOFEN2, was a single-site, dose titration study that was designed to select the maximum tolerated dose for isosorbide dinitrate when coadministered with ibuprofen. Eighteen out of the 19 enrolled subjects tolerated the treatment well, and they completed the study at the highest dose of isosorbide dinitrate applied (80 mg/day). One subject voluntarily decided to reduce the dose of isosorbide dinitrate from 80 mg to 60 mg. The treatment-related adverse events recorded during the study were, for the large majority, episodes of headache that remitted spontaneously in 0.5–1 hour – a known side effect of isosorbide dinitrate. These studies demonstrate that the combination of isosorbide dinitrate and ibuprofen does not lead to pharmacokinetic interactions between the two drugs; they also demonstrate that the combination of isosorbide dinitrate and ibuprofen has optimal tolerability and safety profiles that are similar to those previously reported for isosorbide dinitrate and ibuprofen given alone.
Collapse
Affiliation(s)
- Maria Vittoria Cossu
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Serena Fucile
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Paolo Pellegrino
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Sara Baldelli
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Valeria Cozzi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy
| | - Amedeo Capetti
- Unit of Infectious Diseases, University Hospital "Luigi Sacco", Milan, Italy
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan, Italy ; Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy
| |
Collapse
|
44
|
Falcone G, Perfetti A, Cardinali B, Martelli F. Noncoding RNAs: emerging players in muscular dystrophies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:503634. [PMID: 24729974 PMCID: PMC3960514 DOI: 10.1155/2014/503634] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/17/2014] [Indexed: 12/16/2022]
Abstract
The fascinating world of noncoding RNAs has recently come to light, thanks to the development of powerful sequencing technologies, revealing a variety of RNA molecules playing important regulatory functions in most, if not all, cellular processes. Many noncoding RNAs have been implicated in regulatory networks that are determinant for skeletal muscle differentiation and disease. In this review, we outline the noncoding RNAs involved in physiological mechanisms of myogenesis and those that appear dysregulated in muscle dystrophies, also discussing their potential use as disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Germana Falcone
- Institute of Cell Biology and Neurobiology, National Research Council, 00015 Monterotondo Scalo, Italy
| | - Alessandra Perfetti
- Policlinico San Donato-IRCCS, Molecular Cardiology Laboratory, 20097 San Donato Milanese, Milan, Italy
| | - Beatrice Cardinali
- Institute of Cell Biology and Neurobiology, National Research Council, 00015 Monterotondo Scalo, Italy
| | - Fabio Martelli
- Policlinico San Donato-IRCCS, Molecular Cardiology Laboratory, 20097 San Donato Milanese, Milan, Italy
| |
Collapse
|
45
|
Hashim HZ, Che Abdullah ST, Wan Sulaiman WA, Hoo FK, Basri H. Hunting for a cure: The therapeutic potential of gene therapy in Duchenne muscular dystrophy. Tzu Chi Med J 2014. [DOI: 10.1016/j.tcmj.2014.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
46
|
Shin JY, Dauer WT, Worman HJ. Lamina-associated polypeptide 1: protein interactions and tissue-selective functions. Semin Cell Dev Biol 2014; 29:164-8. [PMID: 24508913 DOI: 10.1016/j.semcdb.2014.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 12/27/2013] [Accepted: 01/28/2014] [Indexed: 01/25/2023]
Abstract
Mutations in genes encoding widely expressed nuclear envelope proteins often lead to diseases that manifest in specific tissues. Lamina-associated polypeptide 1 (LAP1) is an integral protein of the inner nuclear membrane that is expressed in most cells and tissues. Within the nuclear envelope, LAP1 interacts physically with lamins, torsinA and emerin, suggesting it may serve as a key node for transducing signals across the inner nuclear membrane. Indeed, recent in vivo studies in genetically modified mice strongly support functional links between LAP1 and both torsinA (in neurons) and emerin (in muscle). These studies suggest that tissue-selective diseases caused by mutations in genes encoding nuclear envelope proteins may result, at least in part, from the selective disruption of discrete nuclear envelope protein complexes.
Collapse
Affiliation(s)
- Ji-Yeon Shin
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - William T Dauer
- Department of Neurology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
| | - Howard J Worman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
47
|
Rovere-Querini P, Clementi E, Brunelli S. Nitric oxide and muscle repair: multiple actions converging on therapeutic efficacy. Eur J Pharmacol 2013; 730:181-5. [PMID: 24269596 DOI: 10.1016/j.ejphar.2013.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 10/30/2013] [Accepted: 11/08/2013] [Indexed: 12/11/2022]
Abstract
Muscular dystrophies comprise an heterogeneous group of diseases characterised by primary wasting of skeletal muscle, in the most severe forms leading to progressive paralysis and death. Current therapies for these conditions are extremely limited and based on corticosteroids that bear significant side effects. Several studies have proposed possible alternative strategies, ranging from cell and gene therapy to more classical pharmacological approaches. Nitric oxide is a gaseous messenger involved in many mechanisms responsible for preserving muscle function and stimulating muscle repair. We herein review the most recent pre-clinical and clinical findings that open new prospective for the development of nitric oxide as a therapeutic tool for muscular dystrophies.
Collapse
Affiliation(s)
- Patrizia Rovere-Querini
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele University and Scientific Institute, 20132 Milano, Italy
| | - Emilio Clementi
- Scientific Institute IRCCS Eugenio Medea, 23842 Bosisio Parini, Italy; Unit of Clinical Pharmacology, Consiglio Nazionale delle Ricerche, Institute of Neuroscience, Department of Biomedical and Clinical Sciences L. Sacco, University Hospital "Luigi Sacco", Università di Milano, 20157 Milan, Italy
| | - Silvia Brunelli
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele University and Scientific Institute, 20132 Milano, Italy; Department of Health Sciences, Milano-Bicocca University, 20900 Monza, Italy.
| |
Collapse
|