1
|
Riddell D, Harron R, Hildyard J, Wells D, Piercy R. Evaluation of a six-minute walk test in the DE50-MD canine model of Duchenne muscular dystrophy and its effect on blood-borne biomarkers. Wellcome Open Res 2025; 9:681. [PMID: 40292009 PMCID: PMC12022548 DOI: 10.12688/wellcomeopenres.23269.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by mutations in the dystrophin gene resulting in cycles of muscle degeneration, inflammation and regeneration. The 6-minute walk test (6MWT) is a key functional outcome measure for DMD patient clinical trials and has been adapted for use in animal models of the disease. The DE50-MD dog model of DMD closely reflects the DMD patient phenotype prior to loss of ambulation. For pre-clinical trials using this model, functional outcome measures must be established. Methods This longitudinal study compared distance walked in a 6MWT by DE50-MD and WT control dogs and assessed the utility of the 6MWT as a functional biomarker. Dogs underwent two 6MWTs conducted approximately 48-hours apart, at 3, 6, 9, 12, 15 and 18 months of age. In addition, we evaluated the stability of selected blood-borne biomarkers in 12-month old DE50-MD and WT dogs 0, 3, 6, 24 and 48 hours following a 6MWT. Results DE50-MD dogs exhibited significantly shorter 6-minute walk distance (6MWD) than WT dogs at all timepoints (P<0.05), with no difference in 6MWD between the first and second 6MWT. C-C motif chemokine ligand 2 (CCL2), myomesin-3 (MYOM3) and myostatin (MSTN) were biomarkers of the DE50-MD phenotype that remained unchanged in DE50-MD dogs following the 6MWT, while creatine kinase (CK) activity significantly increased 3-hours following the test in DE50-MD dogs but remained unchanged in WT dogs. Conclusions The 6MWT effectively discriminates DE50-MD from WT dogs aged 3-18 months and a single 6MWT is sufficient for future studies. Serum MYOM3, CCL2 and MSTN are good biomarkers of the DE50-MD phenotype that are unaffected by the relatively low level exertion performed in the 6MWT by 12-month-old DE50-MD dogs.
Collapse
Affiliation(s)
- Dominique Riddell
- Comparative Neuromuscular Diseases Laboratory, Clinical Sciences and Services, Royal College Street, London, NW1 0TU, UK
| | - Rachel Harron
- Comparative Neuromuscular Diseases Laboratory, Clinical Sciences and Services, Royal College Street, London, NW1 0TU, UK
| | - John Hildyard
- Comparative Neuromuscular Diseases Laboratory, Clinical Sciences and Services, Royal College Street, London, NW1 0TU, UK
| | - Dominic Wells
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Richard Piercy
- Comparative Neuromuscular Diseases Laboratory, Clinical Sciences and Services, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
2
|
Kiełbowski K, Bakinowska E, Procyk G, Ziętara M, Pawlik A. The Role of MicroRNA in the Pathogenesis of Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:6108. [PMID: 38892293 PMCID: PMC11172814 DOI: 10.3390/ijms25116108] [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: 04/28/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked progressive disorder associated with muscle wasting and degeneration. The disease is caused by mutations in the gene that encodes dystrophin, a protein that links the cytoskeleton with cell membrane proteins. The current treatment methods aim to relieve the symptoms of the disease or partially rescue muscle functionality. However, they are insufficient to suppress disease progression. In recent years, studies have uncovered an important role for non-coding RNAs (ncRNAs) in regulating the progression of numerous diseases. ncRNAs, such as micro-RNAs (miRNAs), bind to their target messenger RNAs (mRNAs) to suppress translation. Understanding the mechanisms involving dysregulated miRNAs can improve diagnosis and suggest novel treatment methods for patients with DMD. This review presents the available evidence on the role of altered expression of miRNAs in the pathogenesis of DMD. We discuss the involvement of these molecules in the processes associated with muscle physiology and DMD-associated cardiomyopathy.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Marta Ziętara
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| |
Collapse
|
3
|
Varvil MS, dos Santos AP. A review on microRNA detection and expression studies in dogs. Front Vet Sci 2023; 10:1261085. [PMID: 37869503 PMCID: PMC10585042 DOI: 10.3389/fvets.2023.1261085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function by post-transcriptional regulation of gene expression. Their stability and abundance in tissue and body fluids makes them promising potential tools for both the diagnosis and prognosis of diseases and attractive therapeutic targets in humans and dogs. Studies of miRNA expression in normal and disease processes in dogs are scarce compared to studies published on miRNA expression in human disease. In this literature review, we identified 461 peer-reviewed papers from database searches using the terms "canine," "dog," "miRNA," and "microRNA"; we screened 244 for inclusion criteria and then included a total of 148 original research peer-reviewed publications relating to specific miRNA expression in canine samples. We found an overlap of miRNA expression changes between the four groups evaluated (normal processes, non-infectious and non-inflammatory conditions, infectious and/or inflammatory conditions, and neoplasia) in 39 miRNAs, 83 miRNAs in three of the four groups, 110 miRNAs in two of the three groups, where 158 miRNAs have only been reported in one of the groups. Additionally, the mechanism of action of these overlapping miRNAs varies depending on the disease process, elucidating a need for characterization of the mechanism of action of each miRNA in each disease process being evaluated. Herein we also draw attention to the lack of standardization of miRNA evaluation, consistency within a single evaluation method, and the need for standardized methods for a direct comparison.
Collapse
Affiliation(s)
- Mara S. Varvil
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
- Department of Veterinary Clinical Sciences, Washington State University, Pullman, WA, United States
| | - Andrea Pires dos Santos
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
4
|
Hanson B, Vorobieva I, Zheng W, Conceição M, Lomonosova Y, Mäger I, Puri PL, El Andaloussi S, Wood MJ, Roberts TC. EV-mediated promotion of myogenic differentiation is dependent on dose, collection medium, and isolation method. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:511-528. [PMID: 37602275 PMCID: PMC10432918 DOI: 10.1016/j.omtn.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 07/11/2023] [Indexed: 08/22/2023]
Abstract
Extracellular vesicles (EVs) have been implicated in the regulation of myogenic differentiation. C2C12 murine myoblast differentiation was reduced following treatment with GW4869 or heparin (to inhibit exosome biogenesis and EV uptake, respectively). Conversely, treatment with C2C12 myotube-conditioned medium enhanced myogenic differentiation. Ultrafiltration-size exclusion liquid chromatography (UF-SEC) was used to isolate EVs and non-EV extracellular protein in parallel from C2C12 myoblast- and myotube-conditioned medium. UF-SEC-purified EVs promoted myogenic differentiation at low doses (≤2 × 108 particles/mL) and were inhibitory at the highest dose tested (2 × 1011 particles/mL). Conversely, extracellular protein fractions had no effect on myogenic differentiation. While the transfer of muscle-enriched miRNAs (myomiRs) has been proposed to mediate the pro-myogenic effects of EVs, we observed that they are scarce in EVs (e.g., 1 copy of miR-133a-3p per 195 EVs). Furthermore, we observed pro-myogenic effects with undifferentiated myoblast-derived EVs, in which myomiR concentrations are even lower, suggestive of a myomiR-independent mechanism underlying the observed pro-myogenic effects. During these investigations we identified technical factors with profound confounding effects on myogenic differentiation. Specifically, co-purification of insulin (a component of Opti-MEM) in non-EV LC fractions and polymer precipitated EV preparations. These findings provide further evidence that polymer-based precipitation techniques should be avoided in EV research.
Collapse
Affiliation(s)
- Britt Hanson
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Ioulia Vorobieva
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Wenyi Zheng
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge SE-141 86, Sweden
| | - Mariana Conceição
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Yulia Lomonosova
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Imre Mäger
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Pier Lorenzo Puri
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA 92037, USA
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge SE-141 86, Sweden
| | - Matthew J.A. Wood
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX3 7TY, UK
| | - Thomas C. Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX3 7TY, UK
| |
Collapse
|
5
|
Yedigaryan L, Sampaolesi M. Extracellular vesicles and Duchenne muscular dystrophy pathology: Modulators of disease progression. Front Physiol 2023; 14:1130063. [PMID: 36891137 PMCID: PMC9987248 DOI: 10.3389/fphys.2023.1130063] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disorder and is considered to be one of the worst forms of inherited muscular dystrophies. DMD occurs as a result of mutations in the dystrophin gene, leading to progressive muscle fiber degradation and weakness. Although DMD pathology has been studied for many years, there are aspects of disease pathogenesis and progression that have not been thoroughly explored yet. The underlying issue with this is that the development of further effective therapies becomes stalled. It is becoming more evident that extracellular vesicles (EVs) may contribute to DMD pathology. EVs are vesicles secreted by cells that exert a multitude of effects via their lipid, protein, and RNA cargo. EV cargo (especially microRNAs) is also said to be a good biomarker for identifying the status of specific pathological processes that occur in dystrophic muscle, such as fibrosis, degeneration, inflammation, adipogenic degeneration, and dilated cardiomyopathy. On the other hand, EVs are becoming more prominent vehicles for custom-engineered cargos. In this review, we will discuss the possible contribution of EVs to DMD pathology, their potential use as biomarkers, and the therapeutic efficacy of both, EV secretion inhibition and custom-engineered cargo delivery.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Chalchat E, Martin V, Charlot K, Bourrilhon C, Baugé S, Bourdon S, Gruel A, Lepetit B, Banzet S, Garcia-Vicencio S, Siracusa J. Circulating microRNA levels after exercise-induced muscle damage and the repeated bout effect. Am J Physiol Regul Integr Comp Physiol 2023; 324:R58-R69. [PMID: 36374177 DOI: 10.1152/ajpregu.00096.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The neuromuscular system can quickly adapt to exercise-induced muscle damage (EIMD), such that it is less affected by subsequent damaging exercise, a phenomenon known as the repeated bout effect (RBE). Circulating muscle-specific microRNAs (myomiRs) may be able to potentially predict the long-lasting maximal voluntary contraction (MVC) torque deficit (>24 h), an indicator of EIMD. We aimed to investigate: 1) how plasma myomiR levels are modified by the RBE and 2) whether plasma myomiRs can predict the long-lasting MVC torque deficit. Nineteen participants performed two identical bouts of loaded downhill walking separated by 2 wk. MVC torque, creatine kinase (CK) activity, myoglobin (Mb) concentration, and myomiR levels were measured before and up to 48 h after exercise. Correlation and multiple regression analyses were performed to assess the ability of these markers to predict the largest MVC torque loss beyond 24 h postexercise. Similar to MVC torque, CK activity, and the Mb concentration, the relative abundance of certain myomiRs (hsa-miR-1-3p, and hsa-miR-133a-3p) was less affected after the second bout of exercise relative to the first bout. The CK activity, Mb concentration, and level of several myomiRs (hsa-miR-1-3p, hsa-miR-133a-3p, and hsa-miR-206) correlated with long-lasting MVC torque loss. Multiple regression showed that the best combination of markers to predict the long-lasting deficit of MVC torque included several myomiRs, Mb, and CK. Certain myomiR levels increased less after exercise bout 2 than after exercise bout 1, indicating the presence of the RBE. The measurement of myomiR levels in combination with Mb concentrations and CK activity could improve the prediction of the long-lasting MVC torque deficit.NEW & NOTEWORTHY The present study is the first to show that plasma muscle-specific microRNA (myomiR) levels can be modified by the repeated bout effect, as their levels increased less after the second exercise bout relative to the first. This study is also the first to suggest that myomiR levels could be used to partially predict maximal voluntary contraction torque loss at 24 h postexercise (i.e., the magnitude of exercise-induced muscle damage). Interestingly, the combined measurement of certain myomiR levels with those of myoglobin and creatine kinase improved the predictive value.
Collapse
Affiliation(s)
- Emeric Chalchat
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,AME2P, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Vincent Martin
- AME2P, Université Clermont Auvergne, Clermont-Ferrand, France.,Institut Universitaire de France, Paris, France
| | - Keyne Charlot
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Cyprien Bourrilhon
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Stéphane Baugé
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Stéphanie Bourdon
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Arnaud Gruel
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Benoit Lepetit
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées, Clamart, France.,INSERM UMRS-MD 1197, Université de Paris-Saclay, Clamart, France
| | - Sebastian Garcia-Vicencio
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France.,Human Motion Analysis, Humanfab, Aix-en-Provence, France
| | - Julien Siracusa
- Département Environnements Opérationnels, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Institut de Recherche Biomédicale des Armées, Bretigny-Sur-Orge, France.,Le Laboratoire de Biologie de l'Exercice pour la Performance et la Santé, Université de Evry, Institut de Recherches Biomédicales des Armées, Université Paris Saclay, Evry, France
| |
Collapse
|
7
|
Riddell DO, Hildyard JCW, Harron RCM, Hornby NL, Wells DJ, Piercy RJ. Serum inflammatory cytokines as disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Dis Model Mech 2022; 15:dmm049394. [PMID: 36444978 PMCID: PMC9789403 DOI: 10.1242/dmm.049394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease, caused by mutations in the dystrophin gene, characterised by cycles of muscle degeneration, inflammation and regeneration. Recently, there has been renewed interest specifically in drugs that ameliorate muscle inflammation in DMD patients. The DE50-MD dog is a model of DMD that closely mimics the human DMD phenotype. We quantified inflammatory proteins in serum from wild-type (WT) and DE50-MD dogs aged 3-18 months to identify biomarkers for future pre-clinical trials. Significantly higher concentrations of C-C motif chemokine ligand 2 (CCL2), granulocyte-macrophage colony-stimulating factor (GM-CSF or CSF2), keratinocyte chemotactic-like (KC-like, homologous to mouse CXCL1), TNFα (or TNF), and interleukins IL2, IL6, IL7, IL8 (CXCL8), IL10, IL15 and IL18 were detected in DE50-MD serum compared to WT serum. Of these, CCL2 best differentiated the two genotypes. The relative level of CCL2 mRNA was greater in the vastus lateralis muscle of DE50-MD dogs than in that of WT dogs, and CCL2 was expressed both within and at the periphery of damaged myofibres. Serum CCL2 concentration was significantly associated with acid phosphatase staining in vastus lateralis biopsy samples in DE50-MD dogs. In conclusion, the serum cytokine profile suggests that inflammation is a feature of the DE50-MD phenotype. Quantification of serum CCL2 in particular is a useful non-invasive biomarker of the DE50-MD phenotype.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Natasha L. Hornby
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Camden, London NW1 0TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, Camden, London NW1 0TU, UK
| |
Collapse
|
8
|
Vu Hong A, Bourg N, Sanatine P, Poupiot J, Charton K, Gicquel E, Massourides E, Spinazzi M, Richard I, Israeli D. Dlk1-Dio3 cluster miRNAs regulate mitochondrial functions in the dystrophic muscle in Duchenne muscular dystrophy. Life Sci Alliance 2022; 6:6/1/e202201506. [PMID: 36265896 PMCID: PMC9585966 DOI: 10.26508/lsa.202201506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by impaired expression of dystrophin. Whereas mitochondrial dysfunction is thought to play an important role in DMD, the mechanism of this dysfunction remains to be clarified. Here we demonstrate that in DMD and other muscular dystrophies, a large number of Dlk1-Dio3 clustered miRNAs (DD-miRNAs) are coordinately up-regulated in regenerating myofibers and in the serum. To characterize the biological effect of this dysregulation, 14 DD-miRNAs were simultaneously overexpressed in vivo in mouse muscle. Transcriptomic analysis revealed highly similar changes between the muscle ectopically overexpressing 14 DD-miRNAs and the mdx diaphragm, with naturally up-regulated DD-miRNAs. Among the commonly dysregulated pathway we found repressed mitochondrial metabolism, and oxidative phosphorylation (OxPhos) in particular. Knocking down the DD-miRNAs in iPS-derived skeletal myotubes resulted in increased OxPhos activities. The data suggest that (1) DD-miRNAs are important mediators of dystrophic changes in DMD muscle, (2) mitochondrial metabolism and OxPhos in particular are targeted in DMD by coordinately up-regulated DD-miRNAs. These findings provide insight into the mechanism of mitochondrial dysfunction in muscular dystrophy.
Collapse
Affiliation(s)
- Ai Vu Hong
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Nathalie Bourg
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Peggy Sanatine
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Jerome Poupiot
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Karine Charton
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - Evelyne Gicquel
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | | | - Marco Spinazzi
- Neuromuscular Reference Center, Department of Neurology, CHU d’Angers, Angers, France,Institute of Neurobiology and Neuropathology CHU d’Angers, Angers, France
| | - Isabelle Richard
- Genethon, Evry, France,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| | - David Israeli
- Genethon, Evry, France .,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare Research Unit UMR_S951, Evry, France
| |
Collapse
|
9
|
Chwalenia K, Oieni J, Zemła J, Lekka M, Ahlskog N, Coenen-Stass AM, McClorey G, Wood MJ, Lomonosova Y, Roberts TC. Exon skipping induces uniform dystrophin rescue with dose-dependent restoration of serum miRNA biomarkers and muscle biophysical properties. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:955-968. [PMID: 36159597 PMCID: PMC9464767 DOI: 10.1016/j.omtn.2022.08.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022]
Abstract
Therapies that restore dystrophin expression are presumed to correct Duchenne muscular dystrophy (DMD), with antisense-mediated exon skipping being the leading approach. Here we aimed to determine whether exon skipping using a peptide-phosphorodiamidate morpholino oligonucleotide (PPMO) conjugate results in dose-dependent restoration of uniform dystrophin localization, together with correction of putative DMD serum and muscle biomarkers. Dystrophin-deficient mdx mice were treated with a PPMO (Pip9b2-PMO) designed to induce Dmd exon 23 skipping at single, ascending intravenous doses (3, 6, or 12 mg/kg) and sacrificed 2 weeks later. Dose-dependent exon skipping and dystrophin protein restoration were observed, with dystrophin uniformly distributed at the sarcolemma of corrected myofibers at all doses. Serum microRNA biomarkers (i.e., miR-1a-3p, miR-133a-3p, miR-206-3p, miR-483-3p) and creatinine kinase levels were restored toward wild-type levels after treatment in a dose-dependent manner. All biomarkers were strongly anti-correlated with both exon skipping level and dystrophin expression. Dystrophin rescue was also strongly positively correlated with muscle stiffness (i.e., Young's modulus) as determined by atomic force microscopy (AFM) nanoindentation assay. These data demonstrate that PPMO-mediated exon skipping generates myofibers with uniform dystrophin expression and that both serum microRNA biomarkers and muscle AFM have potential utility as pharmacodynamic biomarkers of dystrophin restoration therapy in DMD.
Collapse
Affiliation(s)
- Katarzyna Chwalenia
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
| | - Jacopo Oieni
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Joanna Zemła
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland
| | - Nina Ahlskog
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
| | - Anna M.L. Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Graham McClorey
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Matthew J.A. Wood
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX1 3QX, UK
| | - Yulia Lomonosova
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX1 3QX, UK
| | - Thomas C. Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
10
|
Almeida-Becerril T, Rodríguez-Cruz M, Hernández-Cruz SY, Ruiz-Cruz ED, Mendoza CRS, Cárdenas-Conejo A, Escobar-Cedillo RE, Ávila-Moreno F, Aquino-Jarquin G. Natural history of circulating miRNAs in Duchenne disease: Association with muscle injury and metabolic parameters. Acta Neurol Scand 2022; 146:512-524. [PMID: 36000352 DOI: 10.1111/ane.13673] [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: 05/12/2022] [Revised: 06/24/2022] [Accepted: 07/05/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVES This study aimed to evaluate whether the expression of circulating dystromiRs and a group of oxidative stress-related (OS-R) miRNAs is associated with muscle injury and circulating metabolic parameters in Duchenne muscular dystrophy (DMD) patients. METHODS Twenty-four DMD patients were included in this cross-sectional study. Clinical scales to evaluate muscle injury (Vignos, GMFCS, Brooke, and Medical Research Council), enzymatic muscle injury parameters (CPK, ALT, and AST), anthropometry, metabolic indicators, physical activity, serum dystromiRs (miR-1-3p, miR-133a-3p, and miR-206), and OS-R miRNAs (miR-21-5p, miR-31-5p, miR-128-3p, and miR-144-3p) levels were measured in ambulatory and non-ambulatory DMD patients. RESULTS DystromiRs (except miR-1-3p) and miRNAs OS-R levels were lower (p-value <.05) in the non-ambulatory group than the ambulatory group. The expression of those miRNAs correlated with Vignos scale score (For instance, rho = -0.567, p-value <0.05 for miR-21-5p) and with other scales scores of muscle function and strength. CPK, AST, and ALT concentration correlated with expression of all miRNAs (For instance, rho = 0.741, p-value <.05 between miR-206 level and AST concentration). MiR-21-5p level correlated with glucose concentration (rho = -0.369, p-value = .038), and the miR-1-3p level correlated with insulin concentration (rho = 0.343, p-value = .05). CONCLUSIONS Non-ambulatory DMD patients have lower circulating dystromiRs and OS-R miRNAs levels than ambulatory DMD patients. The progressive muscle injury is associated with a decrease in the expression of those miRNAs, evidencing DMD progress. These findings add new information about the natural history of DMD.
Collapse
Affiliation(s)
- Tomas Almeida-Becerril
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), CDMX, Mexico
| | - Maricela Rodríguez-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico
| | - Sthephanie Yannín Hernández-Cruz
- Laboratorio de Nutrición Molecular, Unidad de Investigación Médica en Nutrición, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Mexico City (CDMX), Mexico
| | - Eugenia Dolores Ruiz-Cruz
- Departamento de Genética, Unidad Médica de Alta Especialidad Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional La Raza, IMSS, CDMX, Mexico
| | - Christian Ricardo Sánchez Mendoza
- Departamento de Genética, Unidad Médica de Alta Especialidad Hospital General "Dr. Gaudencio González Garza", Centro Médico Nacional La Raza, IMSS, CDMX, Mexico
| | - Alan Cárdenas-Conejo
- Departamento de Genética Médica, Hospital de Pediatría "Dr. Silvestre Frenk Freund", CMN-Siglo XXI, IMSS, CDMX, Mexico
| | | | - Federico Ávila-Moreno
- Lung Diseases Laboratory 12, Biomedicine Research Unit (UBIMED), Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Guillermo Aquino-Jarquin
- Laboratorio de Investigación en Genómica, Genética y Bioinformática, Hospital Infantil de México "Federico Gómez", CDMX, Mexico
| |
Collapse
|
11
|
Riddell DO, Hildyard JCW, Harron RCM, Wells DJ, Piercy RJ. Longitudinal assessment of blood-borne musculoskeletal disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Wellcome Open Res 2022; 6:354. [PMID: 35600245 PMCID: PMC9111359 DOI: 10.12688/wellcomeopenres.17398.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 01/01/2024] Open
Abstract
Background: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by mutations in the dystrophin gene. Due to their phenotypic similarity to human patients, large animal models are invaluable tools for pre-clinical trials. The DE50-MD dog is a relatively new model of DMD, and carries a therapeutically-tractable mutation lying within the hotspot for human patients, making it especially valuable. Prior to conducting therapeutic trials using this novel animal model, it is essential to establish a panel of viable biomarkers. Methods: We evaluated a panel of blood-borne biomarkers of musculoskeletal disease in the DE50-MD dog. Venous blood samples were obtained monthly throughout an 18-month study period in DE50-MD (N=18) and wild-type (WT) control (N=14) dogs. A panel of potential plasma/serum biomarkers of DMD was measured and their theoretical utility in future clinical trials determined using sample size calculations. Results: Compared to WT dogs, DE50-MD dogs had substantially higher circulating creatine kinase (CK) activities, myomesin-3 (MYOM3), and the dystromiRs miR-1, miR-133a and miR-206, but significantly lower serum myostatin concentrations. An age-associated pattern, similar to that observed in DMD patients, was seen for CK and MYOM3. Sample size calculations suggested that low cohort sizes (N≤3) could be used to detect up to a 50% improvement in DE50-MD results towards WT levels for each biomarker or a combination thereof (via principal component analysis); as few as N=3 animals should enable detection of a 25% improvement using a combined biomarker approach (alpha 0.05, power 0.8). Conclusions: We have established a panel of blood-borne biomarkers that could be used to monitor musculoskeletal disease or response to a therapeutic intervention in the DE50-MD dog using low numbers of animals. The blood biomarker profile closely mimics that of DMD patients, supporting the hypothesis that this DMD model would be suitable for use in pre-clinical trials.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW10TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| |
Collapse
|
12
|
Riddell DO, Hildyard JCW, Harron RCM, Wells DJ, Piercy RJ. Longitudinal assessment of blood-borne musculoskeletal disease biomarkers in the DE50-MD dog model of Duchenne muscular dystrophy. Wellcome Open Res 2022; 6:354. [PMID: 35600245 PMCID: PMC9111359 DOI: 10.12688/wellcomeopenres.17398.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease caused by mutations in the dystrophin gene. Due to their phenotypic similarity to human patients, large animal models are invaluable tools for pre-clinical trials. The DE50-MD dog is a relatively new model of DMD, and carries a therapeutically-tractable mutation lying within the hotspot for human patients, making it especially valuable. Prior to conducting therapeutic trials using this novel animal model, it is essential to establish a panel of viable biomarkers. Methods: We evaluated a panel of blood-borne biomarkers of musculoskeletal disease in the DE50-MD dog. Venous blood samples were obtained monthly throughout an 18-month study period in DE50-MD (N=18) and wild-type (WT) control (N=14) dogs. A panel of potential plasma/serum biomarkers of DMD was measured and their theoretical utility in future clinical trials determined using sample size calculations. Results: Compared to WT dogs, DE50-MD dogs had substantially higher circulating creatine kinase (CK) activities, myomesin-3 (MYOM3), and the dystromiRs miR-1, miR-133a and miR-206, but significantly lower serum myostatin concentrations. An age-associated pattern, similar to that observed in DMD patients, was seen for CK and MYOM3. Sample size calculations suggested that low cohort sizes (N≤3) could be used to detect up to a 50% improvement in DE50-MD results towards WT levels for each biomarker or a combination thereof (via principal component analysis); as few as N=3 animals should enable detection of a 25% improvement using a combined biomarker approach (alpha 0.05, power 0.8). Conclusions: We have established a panel of blood-borne biomarkers that could be used to monitor musculoskeletal disease or response to a therapeutic intervention in the DE50-MD dog using low numbers of animals. The blood biomarker profile closely mimics that of DMD patients, supporting the hypothesis that this DMD model would be suitable for use in pre-clinical trials.
Collapse
Affiliation(s)
- Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - John C. W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Rachel C. M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW10TU, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, NW10TU, UK
| |
Collapse
|
13
|
MicroRNAs in Dystrophinopathy. Int J Mol Sci 2022; 23:ijms23147785. [PMID: 35887128 PMCID: PMC9318410 DOI: 10.3390/ijms23147785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD), which represent the range of dystrophinopathies, account for nearly 80% of muscle dystrophy. DMD and BMD result from the loss of a functional dystrophin protein, and the leading cause of death in these patients is cardiac remodeling and heart failure. The pathogenesis and progression of the more severe form of DMD have been extensively studied and are controlled by many determinants, including microRNAs (miRNAs). The regulatory role of miRNAs in muscle function and the differential miRNA expression in muscular dystrophy indicate the clinical significance of miRNAs. This review discusses the relevant microRNAs as potential biomarkers and therapeutic targets for DMD and DMD cardiomyopathy as examples of dystrophinopathies.
Collapse
|
14
|
Kakouri AC, Koutalianos D, Koutsoulidou A, Oulas A, Tomazou M, Nikolenko N, Turner C, Roos A, Lusakowska A, Janiszewska K, Papadimas GK, Papadopoulos C, Kararizou E, Papanicolaou EZ, Gorman G, Lochmüller H, Spyrou GM, Phylactou LA. Circulating small RNA signatures differentiate accurately the subtypes of muscular dystrophies: small-RNA next-generation sequencing analytics and functional insights. RNA Biol 2022; 19:507-518. [PMID: 35388741 PMCID: PMC8993092 DOI: 10.1080/15476286.2022.2058817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Muscular dystrophies are a group of rare and severe inherited disorders mainly affecting the muscle tissue. Duchene Muscular Dystrophy, Myotonic Dystrophy types 1 and 2, Limb Girdle Muscular Dystrophy and Facioscapulohumeral Muscular Dystrophy are some of the members of this family of disorders. In addition to the current diagnostic tools, there is an increasing interest for the development of novel non-invasive biomarkers for the diagnosis and monitoring of these diseases. miRNAs are small RNA molecules characterized by high stability in blood thus making them ideal biomarker candidates for various diseases. In this study, we present the first genome-wide next-generation small RNA sequencing in serum samples of five different types of muscular dystrophy patients and healthy individuals. We identified many small RNAs including miRNAs, lncRNAs, tRNAs, snoRNAs and snRNAs, that differentially discriminate the muscular dystrophy patients from the healthy individuals. Further analysis of the identified miRNAs showed that some miRNAs can distinguish the muscular dystrophy patients from controls and other miRNAs are specific to the type of muscular dystrophy. Bioinformatics analysis of the target genes for the most significant miRNAs and the biological role of these genes revealed different pathways that the dysregulated miRNAs are involved in each type of muscular dystrophy investigated. In conclusion, this study shows unique signatures of small RNAs circulating in five types of muscular dystrophy patients and provides a useful resource for future studies for the development of miRNA biomarkers in muscular dystrophies and for their involvement in the pathogenesis of the disorders.
Collapse
Affiliation(s)
- Andrea C Kakouri
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Demetris Koutalianos
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Andrie Koutsoulidou
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Anastasis Oulas
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marios Tomazou
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Neurogenetics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Nikoletta Nikolenko
- National Hospital for Neurology and Neurosurgery, Queen Square, University College London Hospitals NHS Foundation Trust, London, UK
| | - Chris Turner
- National Hospital for Neurology and Neurosurgery, Queen Square, University College London Hospitals NHS Foundation Trust, London, UK
| | - Andreas Roos
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany.,Division of Neurology, Department of Medicine, Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Anna Lusakowska
- Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | | | - George K Papadimas
- Department of Neurology, Eginitio hospital, Medical School of Athens, Athens, Greece
| | | | - Evangelia Kararizou
- Department of Neurology, Eginitio hospital, Medical School of Athens, Athens, Greece
| | | | - Grainne Gorman
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, University of Newcastle, Newcastle, UK
| | - Hanns Lochmüller
- Division of Neurology, Department of Medicine, Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Centro Nacional de AnálisisGenómico, Center for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology (Bist), Barcelona, Spain
| | - George M Spyrou
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Leonidas A Phylactou
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This review highlights the key studies investigating various types of biomarkers in Duchenne muscular dystrophy (DMD). RECENT FINDINGS Several proteomic and metabolomic studies have been undertaken in both human DMD patients and animal models of DMD that have identified potential biomarkers in DMD. Although there have been a number of proteomic and metabolomic studies that have identified various potential biomarkers in DMD, more definitive studies still need to be undertaken in DMD patients to firmly correlate these biomarkers with diagnosis, disease progression, and monitoring the effects of novel treatment strategies being developed.
Collapse
Affiliation(s)
- Theo Lee-Gannon
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xuan Jiang
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tara C Tassin
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pradeep P A Mammen
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
16
|
Gartz M, Beatka M, Prom MJ, Strande JL, Lawlor MW. Cardiomyocyte-produced miR-339-5p mediates pathology in Duchenne muscular dystrophy cardiomyopathy. Hum Mol Genet 2021; 30:2347-2361. [PMID: 34270708 PMCID: PMC8600005 DOI: 10.1093/hmg/ddab199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/19/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by severe, progressive muscle wasting. Cardiomyopathy has emerged as a leading cause of death in patients with DMD. The mechanisms contributing to DMD cardiac disease remain under investigation and specific therapies available are lacking. Our prior work has shown that DMD-iPSC-derived cardiomyocytes (DMD-iCMs) are vulnerable to oxidative stress injury and chronic exposure to DMD-secreted exosomes impaired the cell's ability to protect against stress. In this study, we sought to examine a mechanism by which DMD cardiac exosomes impair cellular response through altering important stress-responsive genes in the recipient cells. Here, we report that DMD-iCMs secrete exosomes containing altered microRNA (miR) profiles in comparison to healthy controls. In particular, miR-339-5p was upregulated in DMD-iCMs, DMD exosomes and mdx mouse cardiac tissue. Restoring dystrophin in DMD-iCMs improved the cellular response to stress and was associated with downregulation of miR-339-5p, suggesting that it is disease-specific. Knockdown of miR-339-5p was associated with increased expression of MDM2, GSK3A and MAP2K3, which are genes involved in important stress-responsive signaling pathways. Finally, knockdown of miR-339-5p led to mitochondrial protection and a reduction in cell death in DMD-iCMs, indicating miR-339-5p is involved in direct modulation of stress-responsiveness. Together, these findings identify a potential mechanism by which exosomal miR-339-5p may be modulating cell signaling pathways that are important for robust stress responses. Additionally, these exosomal miRs may provide important disease-specific targets for future therapeutic advancements for the management and diagnosis of DMD cardiomyopathy.
Collapse
Affiliation(s)
- Melanie Gartz
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Margaret Beatka
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mariah J Prom
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer L Strande
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael W Lawlor
- Department of Cell Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
17
|
Yedigaryan L, Sampaolesi M. Therapeutic Implications of miRNAs for Muscle-Wasting Conditions. Cells 2021; 10:cells10113035. [PMID: 34831256 PMCID: PMC8616481 DOI: 10.3390/cells10113035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that are mainly involved in translational repression by binding to specific messenger RNAs. Recently, miRNAs have emerged as biomarkers, relevant for a multitude of pathophysiological conditions, and cells can selectively sort miRNAs into extracellular vesicles for paracrine and endocrine effects. In the overall context of muscle-wasting conditions, a multitude of miRNAs has been implied as being responsible for the typical dysregulation of anabolic and catabolic pathways. In general, chronic muscle disorders are associated with the main characteristic of a substantial loss in muscle mass. Muscular dystrophies (MDs) are a group of genetic diseases that cause muscle weakness and degeneration. Typically, MDs are caused by mutations in those genes responsible for upholding the integrity of muscle structure and function. Recently, the dysregulation of miRNA levels in such pathological conditions has been reported. This revelation is imperative for both MDs and other muscle-wasting conditions, such as sarcopenia and cancer cachexia. The expression levels of miRNAs have immense potential for use as potential diagnostic, prognostic and therapeutic biomarkers. Understanding the role of miRNAs in muscle-wasting conditions may lead to the development of novel strategies for the improvement of patient management.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
18
|
Willner EC, Galan HL, Cuneo BF, Hoffman HA, Neltner B, Schuchardt EL, Karimpour-Fard A, Miyamoto SD, Sucharov CC. Amniotic fluid microRNA profiles in twin-twin transfusion syndrome with and without severe recipient cardiomyopathy. Am J Obstet Gynecol 2021; 225:439.e1-439.e10. [PMID: 34153234 DOI: 10.1016/j.ajog.2021.06.066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Twin-twin transfusion syndrome presents many challenges for clinicians, and the optimal means of identifying pregnancies that will benefit most from intervention is controversial. There is currently no clinically available biomarker to detect twin-twin transfusion syndrome or to stratify cases based on the risk factors. microRNAs are small RNAs that regulate gene expression and are biomarkers for various disease processes, including adult and pediatric heart failure. To date, no studies have investigated amniotic fluid microRNAs as biomarkers for disease severity, specifically for severe recipient cardiomyopathy in twin-twin transfusion syndrome cases. OBJECTIVE This study aimed to assess whether amniotic fluid microRNAs could be useful as biomarkers to identify pregnancies at greatest risk for severe recipient cardiomyopathy associated with twin-twin transfusion syndrome. STUDY DESIGN Amniotic fluid was collected at the time of amnioreduction or selective fetoscopic laser photocoagulation from monochorionic diamniotic twin pregnancies with twin-twin transfusion syndrome at any stage. Fetal echocardiography was performed on all twins before the procedure, and severe cardiomyopathy was defined as a right ventricular myocardial performance index of the recipient fetus of >4 Z-scores. microRNA was extracted from the amniotic fluid samples and analyzed using an array panel assessing 379 microRNAs (TaqMan Open Array, ThermoFisher). Student t tests were performed to determine significant differences in microRNA expression between pregnancies with severe recipient cardiomyopathy and those with preserved cardiac function. A stringent q value of <.0025 was used to determine differential microRNA expression. Random forest plots identified the top 3 microRNAs that separated the 2 groups, and hierarchical cluster analysis was used to determine if these microRNAs properly segregated the samples according to their clinical groups. RESULTS A total of 14 amniotic fluid samples from pregnancies with twin-twin transfusion syndrome with severe cardiomyopathy were compared with samples from 12 twin-twin transfusion syndrome control cases with preserved cardiac function. A total of 110 microRNAs were identified in the amniotic fluid samples. Twenty microRNAs were differentially expressed, and the top 3 differentiating microRNAs were hsa-miR-200c-3p, hsa-miR-17-5p, and hsa-miR-539-5p. Hierarchical cluster analysis based on these top 3 microRNAs showed a strong ability to differentiate severe cardiomyopathy cases from controls. The top 3 microRNAs were used to investigate the sensitivity and specificity of these microRNAs to differentiate between the 2 groups with a receiver operating characteristic curve demonstrating sensitivity and specificity of 80.8%. All 20 differentially expressed microRNAs were down-regulated in the group with severe cardiomyopathy. CONCLUSION Amniotic fluid microRNAs demonstrated differential expression between twin-twin transfusion syndrome recipient fetuses with severe cardiomyopathy and those without and have the potential to be important biomarkers of disease severity in this population.
Collapse
Affiliation(s)
- Emily C Willner
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO
| | - Henry L Galan
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO
| | - Bettina F Cuneo
- Division of Cardiology, Department of Pediatrics, Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO
| | - Hilary A Hoffman
- Colorado Fetal Care Center, Children's Hospital Colorado, Aurora, CO
| | - Bonnie Neltner
- Division of Cardiology, University of Colorado, Aurora, CO
| | - Eleanor L Schuchardt
- Department of Pediatrics, Children's Hospital Colorado, Aurora, CO; Division of Cardiology, Department of Pediatrics, University of California San Diego and Rady Children's Hospital, San Diego, CA
| | | | - Shelley D Miyamoto
- Department of Pediatrics, Children's Hospital Colorado, Aurora, CO; Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado, Aurora, CO.
| |
Collapse
|
19
|
Vu Hong A, Sanson M, Richard I, Israeli D. A revised model for mitochondrial dysfunction in Duchenne muscular dystrophy. Eur J Transl Myol 2021; 31. [PMID: 34533019 PMCID: PMC8495359 DOI: 10.4081/ejtm.2021.10012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/12/2021] [Indexed: 12/27/2022] Open
Abstract
We recently identified a signaling pathway that links the upregulation of miR-379 with a mitochondrial response in dystrophic muscle. In the present commentary, we explain the significance that this pathway may have in mitochondrial dysfunction in Duchenne muscular dystrophy (DMD). We identified the upregulation of miR-379 in the serum and muscles of DMD animal models and patients. We found that miR-379 is one of very few miRNAs whose expression was normalized in DMD patients treated with glucocorticoid. We identified EIF4G2 as a miR-379 target, which may promote mitochondrial oxidative phosphorylation (OxPhos) in the skeletal muscle. We found enriched EIF4G2 expression in oxidative fibers, and identified the mitochondrial ATP synthase subunit DAPIT as a translational target of EIF4G2. The identified signaling cascade, which comprises miR-379, EIF4G2 and DAPIT, may link the glucocorticoid treatment in DMD to a recovered mitochondrial ATP synthesis rate. We propose an updated model of mitochondrial dysfunction in DMD.
Collapse
Affiliation(s)
- Ai Vu Hong
- Genethon, Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR-S951, Evry.
| | - Mathilde Sanson
- Genethon, Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR-S951, Evry.
| | - Isabelle Richard
- Genethon, Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR-S951, Evry.
| | - David Israeli
- Genethon, Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR-S951, Evry.
| |
Collapse
|
20
|
Tsonaka R, Signorelli M, Sabir E, Seyer A, Hettne K, Aartsma-Rus A, Spitali P. Longitudinal metabolomic analysis of plasma enables modeling disease progression in Duchenne muscular dystrophy mouse models. Hum Mol Genet 2021; 29:745-755. [PMID: 32025735 PMCID: PMC7104681 DOI: 10.1093/hmg/ddz309] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy is a severe pediatric neuromuscular disorder caused by the lack of dystrophin. Identification of biomarkers is needed to support and accelerate drug development. Alterations of metabolites levels in muscle and plasma have been reported in pre-clinical and clinical cross-sectional comparisons. We present here a 7-month longitudinal study comparing plasma metabolomic data in wild-type and mdx mice. A mass spectrometry approach was used to study metabolites in up to five time points per mouse at 6, 12, 18, 24 and 30 weeks of age, providing an unprecedented in depth view of disease trajectories. A total of 106 metabolites were studied. We report a signature of 31 metabolites able to discriminate between healthy and disease at various stages of the disease, covering the acute phase of muscle degeneration and regeneration up to the deteriorating phase. We show how metabolites related to energy production and chachexia (e.g. glutamine) are affected in mdx mice plasma over time. We further show how the signature is connected to molecular targets of nutraceuticals and pharmaceutical compounds currently in development as well as to the nitric oxide synthase pathway (e.g. arginine and citrulline). Finally, we evaluate the signature in a second longitudinal study in three independent mouse models carrying 0, 1 or 2 functional copies of the dystrophin paralog utrophin. In conclusion, we report an in-depth metabolomic signature covering previously identified associations and new associations, which enables drug developers to peripherally assess the effect of drugs on the metabolic status of dystrophic mice.
Collapse
Affiliation(s)
- Roula Tsonaka
- Biomedical Data Sciences, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | - Mirko Signorelli
- Biomedical Data Sciences, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | - Ekrem Sabir
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | | | - Kristina Hettne
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, The Netherlands
| |
Collapse
|
21
|
Amor F, Vu Hong A, Corre G, Sanson M, Suel L, Blaie S, Servais L, Voit T, Richard I, Israeli D. Cholesterol metabolism is a potential therapeutic target in Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2021; 12:677-693. [PMID: 34037326 PMCID: PMC8200436 DOI: 10.1002/jcsm.12708] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a lethal muscle disease detected in approximately 1:5000 male births. DMD is caused by mutations in the DMD gene, encoding a critical protein that links the cytoskeleton and the extracellular matrix in skeletal and cardiac muscles. The primary consequence of the disrupted link between the extracellular matrix and the myofibre actin cytoskeleton is thought to involve sarcolemma destabilization, perturbation of Ca2+ homeostasis, activation of proteases, mitochondrial damage, and tissue degeneration. A recently emphasized secondary aspect of the dystrophic process is a progressive metabolic change of the dystrophic tissue; however, the mechanism and nature of the metabolic dysregulation are yet poorly understood. In this study, we characterized a molecular mechanism of metabolic perturbation in DMD. METHODS We sequenced plasma miRNA in a DMD cohort, comprising 54 DMD patients treated or not by glucocorticoid, compared with 27 healthy controls, in three groups of the ages of 4-8, 8-12, and 12-20 years. We developed an original approach for the biological interpretation of miRNA dysregulation and produced a novel hypothesis concerning metabolic perturbation in DMD. We used the mdx mouse model for DMD for the investigation of this hypothesis. RESULTS We identified 96 dysregulated miRNAs (adjusted P-value <0.1), of which 74 were up-regulated and 22 were down-regulated in DMD. We confirmed the dysregulation in DMD of Dystro-miRs, Cardio-miRs, and a large number of the DLK1-DIO3 miRNAs. We also identified numerous dysregulated miRNAs yet unreported in DMD. Bioinformatics analysis of both target and host genes for dysregulated miRNAs predicted that lipid metabolism might be a critical metabolic perturbation in DMD. Investigation of skeletal muscles of the mdx mouse uncovered dysregulation of transcription factors of cholesterol and fatty acid metabolism (SREBP-1 and SREBP-2), perturbation of the mevalonate pathway, and the accumulation of cholesterol in the dystrophic muscles. Elevated cholesterol level was also found in muscle biopsies of DMD patients. Treatment of mdx mice with Simvastatin, a cholesterol-reducing agent, normalized these perturbations and partially restored the dystrophic parameters. CONCLUSIONS This investigation supports that cholesterol metabolism and the mevalonate pathway are potential therapeutic targets in DMD.
Collapse
Affiliation(s)
- Fatima Amor
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | - Ai Vu Hong
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | - Guillaume Corre
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | - Mathilde Sanson
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | - Laurence Suel
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | | | - Laurent Servais
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, UK & Division of Child Neurology, Centre de Référence des Maladies Neuromusculaires, Department of PaediatricsUniversity Hospital of Liège & University of LiègeLiègeBelgium
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Isabelle Richard
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| | - David Israeli
- GénéthonEvryFrance
- Université Paris‐Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951EvryFrance
| |
Collapse
|
22
|
Daza KR, Velez-Irizarry D, Casiró S, Steibel JP, Raney NE, Bates RO, Ernst CW. Integrated Genome-Wide Analysis of MicroRNA Expression Quantitative Trait Loci in Pig Longissimus Dorsi Muscle. Front Genet 2021; 12:644091. [PMID: 33859669 PMCID: PMC8042294 DOI: 10.3389/fgene.2021.644091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/24/2021] [Indexed: 01/19/2023] Open
Abstract
Determining mechanisms regulating complex traits in pigs is essential to improve the production efficiency of this globally important protein source. MicroRNAs (miRNAs) are a class of non-coding RNAs known to post-transcriptionally regulate gene expression affecting numerous phenotypes, including those important to the pig industry. To facilitate a more comprehensive understanding of the regulatory mechanisms controlling growth, carcass composition, and meat quality phenotypes in pigs, we integrated miRNA and gene expression data from longissimus dorsi muscle samples with genotypic and phenotypic data from the same animals. We identified 23 miRNA expression Quantitative Trait Loci (miR-eQTL) at the genome-wide level and examined their potential effects on these important production phenotypes through miRNA target prediction, correlation, and colocalization analyses. One miR-eQTL miRNA, miR-874, has target genes that colocalize with phenotypic QTL for 12 production traits across the genome including backfat thickness, dressing percentage, muscle pH at 24 h post-mortem, and cook yield. The results of our study reveal genomic regions underlying variation in miRNA expression and identify miRNAs and genes for future validation of their regulatory effects on traits of economic importance to the global pig industry.
Collapse
Affiliation(s)
- Kaitlyn R Daza
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Deborah Velez-Irizarry
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Sebastian Casiró
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Juan P Steibel
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Nancy E Raney
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Ronald O Bates
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Catherine W Ernst
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
23
|
Carraro U, Yablonka-Reuveni Z. Translational research on Myology and Mobility Medicine: 2021 semi-virtual PDM3 from Thermae of Euganean Hills, May 26 - 29, 2021. Eur J Transl Myol 2021; 31:9743. [PMID: 33733717 PMCID: PMC8056169 DOI: 10.4081/ejtm.2021.9743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
On 19-21 November 2020, the meeting of the 30 years of the Padova Muscle Days was virtually held while the SARS-CoV-2 epidemic was hitting the world after a seemingly quiet summer. During the 2020-2021 winter, the epidemic is still active, despite the start of vaccinations. The organizers hope to hold the 2021 Padua Days on Myology and Mobility Medicine in a semi-virtual form (2021 S-V PDM3) from May 26 to May 29 at the Thermae of Euganean Hills, Padova, Italy. Here the program and the Collection of Abstracts are presented. Despite numerous world problems, the number of submitted/selected presentations (lectures and oral presentations) has increased, prompting the organizers to extend the program to four dense days.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences of the University of Padova, Italy; CIR-Myo - Myology Centre, University of Padova, Italy; A-C Mioni-Carraro Foundation for Translational Myology, Padova.
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
24
|
Singh GB, Cowan DB, Wang DZ. Tiny Regulators of Massive Tissue: MicroRNAs in Skeletal Muscle Development, Myopathies, and Cancer Cachexia. Front Oncol 2020; 10:598964. [PMID: 33330096 PMCID: PMC7719840 DOI: 10.3389/fonc.2020.598964] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscles are the largest tissues in our body and the physiological function of muscle is essential to every aspect of life. The regulation of development, homeostasis, and metabolism is critical for the proper functioning of skeletal muscle. Consequently, understanding the processes involved in the regulation of myogenesis is of great interest. Non-coding RNAs especially microRNAs (miRNAs) are important regulators of gene expression and function. MiRNAs are small (~22 nucleotides long) noncoding RNAs known to negatively regulate target gene expression post-transcriptionally and are abundantly expressed in skeletal muscle. Gain- and loss-of function studies have revealed important roles of this class of small molecules in muscle biology and disease. In this review, we summarize the latest research that explores the role of miRNAs in skeletal muscle development, gene expression, and function as well as in muscle disorders like sarcopenia and Duchenne muscular dystrophy (DMD). Continuing with the theme of the current review series, we also briefly discuss the role of miRNAs in cancer cachexia.
Collapse
Affiliation(s)
- Gurinder Bir Singh
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Douglas B Cowan
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
25
|
Heier CR, Zhang A, Nguyen NY, Tully CB, Panigrahi A, Gordish-Dressman H, Pandey SN, Guglieri M, Ryan MM, Clemens PR, Thangarajh M, Webster R, Smith EC, Connolly AM, McDonald CM, Karachunski P, Tulinius M, Harper A, Mah JK, Fiorillo AA, Chen YW. Multi-Omics Identifies Circulating miRNA and Protein Biomarkers for Facioscapulohumeral Dystrophy. J Pers Med 2020; 10:jpm10040236. [PMID: 33228131 PMCID: PMC7711540 DOI: 10.3390/jpm10040236] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
The development of therapeutics for muscle diseases such as facioscapulohumeral dystrophy (FSHD) is impeded by a lack of objective, minimally invasive biomarkers. Here we identify circulating miRNAs and proteins that are dysregulated in early-onset FSHD patients to develop blood-based molecular biomarkers. Plasma samples from clinically characterized individuals with early-onset FSHD provide a discovery group and are compared to healthy control volunteers. Low-density quantitative polymerase chain reaction (PCR)-based arrays identify 19 candidate miRNAs, while mass spectrometry proteomic analysis identifies 13 candidate proteins. Bioinformatic analysis of chromatin immunoprecipitation (ChIP)-seq data shows that the FSHD-dysregulated DUX4 transcription factor binds to regulatory regions of several candidate miRNAs. This panel of miRNAs also shows ChIP signatures consistent with regulation by additional transcription factors which are up-regulated in FSHD (FOS, EGR1, MYC, and YY1). Validation studies in a separate group of patients with FSHD show consistent up-regulation of miR-100, miR-103, miR-146b, miR-29b, miR-34a, miR-454, miR-505, and miR-576. An increase in the expression of S100A8 protein, an inflammatory regulatory factor and subunit of calprotectin, is validated by Enzyme-Linked Immunosorbent Assay (ELISA). Bioinformatic analyses of proteomics and miRNA data further support a model of calprotectin and toll-like receptor 4 (TLR4) pathway dysregulation in FSHD. Moving forward, this panel of miRNAs, along with S100A8 and calprotectin, merit further investigation as monitoring and pharmacodynamic biomarkers for FSHD.
Collapse
Affiliation(s)
- Christopher R. Heier
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Correspondence: (C.R.H.); (Y.-W.C.)
| | - Aiping Zhang
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Nhu Y Nguyen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Aswini Panigrahi
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Heather Gordish-Dressman
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Sachchida Nand Pandey
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | | | - Monique M. Ryan
- The Royal Children’s Hospital, Melbourne University, Parkville, Victoria 3052, Australia;
| | - Paula R. Clemens
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Mathula Thangarajh
- Department of Neurology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA;
| | | | - Edward C. Smith
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27705, USA;
| | - Anne M. Connolly
- Nationwide Children’s Hospital, The Ohio State University, Columbus, OH 43205, USA;
| | - Craig M. McDonald
- Department of Physical Medicine and Rehabilitation, University of California at Davis Medical Center, Sacramento, CA 95817, USA;
| | - Peter Karachunski
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Mar Tulinius
- Department of Pediatrics, Gothenburg University, Queen Silvia Children’s Hospital, 41685 Göteborg, Sweden;
| | - Amy Harper
- Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Jean K. Mah
- Deparment of Pediatrics and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, T2N T3B, Calgary, AB 6A81N4, Canada;
| | - Alyson A. Fiorillo
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; (H.G.-D.); (A.A.F.)
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20010, USA; (A.Z.); (N.Y.N.); (C.B.T.); (A.P.); (S.N.P.)
- Correspondence: (C.R.H.); (Y.-W.C.)
| | | |
Collapse
|
26
|
Carraro U. 30 Years of Translational Mobility Medicine: 2020 Padua Muscle Days go virtual from Euganean Hills, November 19th to 21st. Eur J Transl Myol 2020; 30:9437. [PMID: 33520146 PMCID: PMC7844408 DOI: 10.4081/ejtm.2020.9437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
In the autumn of 2019, the 2020 Padua Muscle Days (PMDs) were planned to be held from March 18 to March 21, 2020. The program listed Scientific Sessions to occur over three full days at either Padova University or the Hotel Augustus on Euganei Hills (Padova), Italy. Abruptly, however, in early January the Coronavirus COVID-19 outbreak started in China and changed the world perspectives. In Italy, the epidemia had the first Italian cases and victims in an area south of Milan and in a Village of the Euganei Hills (Vo Euganeo, Padova). Thus, it was a mandatory decision to post-pone the PMDs meeting to 19-21 November, 2020. Luckily, almost all chairs, speakers, and attendees accepted the decision and have assured their presence in late November by long-distance communications. Thus, the Collection of Abstracts were e-published in 30(1) 2020 Issue of the European Journal of Translational Myology (EJTM) together with the many EJTM Communications submitted by speakers and attendees of the 2020 PMDs Here we add a few new entries and the detailed Program of the 2020 Virtual PMDs to be organized November 19-21, 2020 from the Hotel Petrarca of Euganei Hills (Padova), Italy. The Program of the 2020 Virtual PMDs ends with invitation by Zipora Yablonka-Reuveni and myself to the 2021 (Virtual) Padua Muscle Days, March 25-27, Euganei Hills (Padova), Italy.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences, University of Padova, Italy.,Armando & Carmela Mioni-Carraro Foundation for Translational Myology Padova, Italy
| |
Collapse
|
27
|
Brusa R, Magri F, Bresolin N, Comi GP, Corti S. Noncoding RNAs in Duchenne and Becker muscular dystrophies: role in pathogenesis and future prognostic and therapeutic perspectives. Cell Mol Life Sci 2020; 77:4299-4313. [PMID: 32350552 PMCID: PMC11105074 DOI: 10.1007/s00018-020-03537-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Noncoding RNAs (ncRNAs), such as miRNAs and long noncoding RNAs, are key regulators of gene expression at the post-transcriptional level and represent promising therapeutic targets and biomarkers for several human diseases, including Duchenne and Becker muscular dystrophies (DMD/BMD). A role for ncRNAs in the pathogenesis of muscular dystrophies has been suggested, even if it is still incompletely understood. Here, we discuss current progress leading towards the clinical utility of ncRNAs for DMD/BMD. Long and short noncoding RNAs are differentially expressed in DMD/BMD and have a mechanism of action via targeting mRNAs. A subset of muscle-enriched miRNAs, the so-called myomiRs (miR-1, miR-133, and miR-206), are increased in the serum of patients with DMD and in dystrophin-defective animal models. Interestingly, myomiRs might be used as biomarkers, given that their levels can be corrected after dystrophin restoration in dystrophic mice. Remarkably, further evidence demonstrates that ncRNAs also play a role in dystrophin expression; thus, their modulations might represent a potential therapeutic strategy with the aim of upregulating the dystrophin protein in combination with other oligonucleotides/gene therapy approaches.
Collapse
Affiliation(s)
- Roberta Brusa
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Magri
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nereo Bresolin
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.
| |
Collapse
|
28
|
Koutsoulidou A, Phylactou LA. Circulating Biomarkers in Muscular Dystrophies: Disease and Therapy Monitoring. Mol Ther Methods Clin Dev 2020; 18:230-239. [PMID: 32637452 PMCID: PMC7327849 DOI: 10.1016/j.omtm.2020.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Muscular dystrophies are a group of inherited disorders that primarily affect the muscle tissues. Across the muscular dystrophies, symptoms commonly compromise the quality of life in all areas of functioning. It is well noted that muscular dystrophies need reliable and measurable biomarkers that will monitor the progress of the disease and evaluate the potential therapeutic approaches. In this review, we analyze the current findings regarding the development of blood-based circulating biomarkers for different types of muscular dystrophies. We emphasize those muscular dystrophies that gained particular interest for the development of biomarkers, including Duchenne muscular dystrophy, Becker muscular dystrophy, myotonic dystrophy types 1 and 2, Ullrich congenital muscular dystrophy, congenital muscular dystrophy type 1A, Facioscapulohumeral muscular dystrophy, and limb-girdle muscular dystrophy types 2A, 2B, 2C, and 2D, recently renamed as limb-girdle muscular dystrophy R1 calpain3-related, R2 dysferlin-related, R5 γ-sarcoglycan-related, and R3 α-sarcoglycan-related. This review highlights the up-to-date progress in the development of biomarkers at the level of proteins, lipids, and metabolites, as well as microRNAs (miRNAs) that currently are the main potential biomarker candidates in muscular dystrophies.
Collapse
Affiliation(s)
- Andrie Koutsoulidou
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology and Genetics, PO Box 23462, 1683 Nicosia, Cyprus.,Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, PO Box 23462, 1683 Nicosia, Cyprus
| | - Leonidas A Phylactou
- Department of Molecular Genetics, Function & Therapy, The Cyprus Institute of Neurology and Genetics, PO Box 23462, 1683 Nicosia, Cyprus.,Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, PO Box 23462, 1683 Nicosia, Cyprus
| |
Collapse
|
29
|
Comprehensive Analysis of Long Non-coding RNA-Associated Competing Endogenous RNA Network in Duchenne Muscular Dystrophy. Interdiscip Sci 2020; 12:447-460. [PMID: 32876881 DOI: 10.1007/s12539-020-00388-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is one of the most severe neuromuscular disorders. Long non-coding RNAs (lncRNAs) are a group of non-coding transcripts, which could regulate messenger RNA (mRNA) by binding the mutual miRNAs, thus acting as competing endogenous RNAs (ceRNAs). So far, the role of lncRNA in DMD pathogenesis remains unclear. In the current study, expression profile from a total of 33 DMD patients and 12 healthy people were downloaded from Gene Expression Omnibus (GEO) database (GSE38417 and GSE109178). Differentially expressed (DE) lncRNAs were discovered and targeted mRNAs were predicted. The ceRNA network of lncRNAs-miRNAs-mRNAs was then constructed. Genome Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of the putative mRNAs in the ceRNA network were performed through Database for Annotation, Visualization and Integration Discovery (DAVID) website. Topological property of the network was analyzed using Cytoscape to disclose the hub lncRNAs. According to our assessments, 19 common DElncRNAs and 846 common DEmRNAs were identified in DMD compared to controls. The created ceRNA network contained 6 lncRNA nodes, 69 mRNA nodes, 27 miRNA nodes and 102 edges, while four hub lncRNAs (XIST, AL132709, LINC00310, ALDH1L1-AS2) were uncovered. In conclusion, our latest bioinformatic analysis demonstrated that lncRNA is likely involved in DMD. This work highlights the importance of lncRNA and provides new insights for exploring the molecular mechanism of DMD. The created ceRNA network contained 6 lncRNA nodes, 69 mRNA nodes, 27 miRNA nodes and 102 edges, while four hub lncRNAs (XIST, AL132709, LINC00310, ALDH1L1-AS2) were uncovered. Remarkably, KEGG analysis indicated that targeted mRNAs in the network were mainly enriched in "microRNAs in cancer" and "proteoglycans in cancer". Our study may offer novel perspectives on the pathogenesis of DMD from the point of lncRNAs. This work might be also conducive for exploring the molecular mechanism of increased incidence of tumorigenesis reported in DMD patients and experimental models.
Collapse
|
30
|
Koller D, Kubinyi E, Elek Z, Nemeth H, Miklosi A, Sasvari-Szekely M, Ronai Z. The molecular effect of a polymorphic microRNA binding site of Wolfram syndrome 1 gene in dogs. BMC Genet 2020; 21:82. [PMID: 32723293 PMCID: PMC7390163 DOI: 10.1186/s12863-020-00879-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/29/2020] [Indexed: 11/10/2022] Open
Abstract
Background Although the molecular function of wolframin remains unclear, the lack of this protein is known to cause stress in the endoplasmic reticulum. Some variants in the Wolfram Syndrome 1 gene (WFS1) were associated with various neuropsychiatric disorders in humans, such as aggressiveness, impulsivity and anxiety. Results Here we present an in silico study predicting a single nucleotide polymorphism (rs852850348) in the canine WFS1 gene which was verified by direct sequencing and was genotyped by a PCR-based technique. We found that the rs852850348 polymorphism is located in a putative microRNA (cfa-miR-8834a and cfa-miR-1838) binding site. Therefore, the molecular effect of allelic variants was studied in a luciferase reporter system that allowed assessing gene expression. We demonstrated that the variant reduced the activity of the reporter protein expression in an allele-specific manner. Additionally, we performed a behavioral experiment and investigated the association with this locus to different performance in this test. Association was found between food possessivity and the studied WFS1 gene polymorphism in the Border collie breed. Conclusions Based on our findings, the rs852850348 locus might contribute to the genetic risk of possessivity behavior of dogs in at least one breed and might influence the regulation of wolframin expression.
Collapse
Affiliation(s)
- Dora Koller
- Comparative Ethology Research Group, MTA-ELTE, Budapest, Hungary. .,Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary. .,Department of Ethology, ELTE Eötvös Loránd University, Budapest, Hungary.
| | - Eniko Kubinyi
- Comparative Ethology Research Group, MTA-ELTE, Budapest, Hungary.,Department of Ethology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsanna Elek
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Helga Nemeth
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Adam Miklosi
- Comparative Ethology Research Group, MTA-ELTE, Budapest, Hungary.,Department of Ethology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Maria Sasvari-Szekely
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Zsolt Ronai
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
31
|
Sanson M, Vu Hong A, Massourides E, Bourg N, Suel L, Amor F, Corre G, Bénit P, Barthélémy I, Blot S, Bigot A, Pinset C, Rustin P, Servais L, Voit T, Richard I, Israeli D. miR-379 links glucocorticoid treatment with mitochondrial response in Duchenne muscular dystrophy. Sci Rep 2020; 10:9139. [PMID: 32499563 PMCID: PMC7272451 DOI: 10.1038/s41598-020-66016-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal muscle disorder, caused by mutations in the DMD gene and affects approximately 1:5000-6000 male births. In this report, we identified dysregulation of members of the Dlk1-Dio3 miRNA cluster in muscle biopsies of the GRMD dog model. Of these, we selected miR-379 for a detailed investigation because its expression is high in the muscle, and is known to be responsive to glucocorticoid, a class of anti-inflammatory drugs commonly used in DMD patients. Bioinformatics analysis predicts that miR-379 targets EIF4G2, a translational factor, which is involved in the control of mitochondrial metabolic maturation. We confirmed in myoblasts that EIF4G2 is a direct target of miR-379, and identified the DAPIT mitochondrial protein as a translational target of EIF4G2. Knocking down DAPIT in skeletal myotubes resulted in reduced ATP synthesis and myogenic differentiation. We also demonstrated that this pathway is GC-responsive since treating mice with dexamethasone resulted in reduced muscle expression of miR-379 and increased expression of EIF4G2 and DAPIT. Furthermore, miR-379 seric level, which is also elevated in the plasma of DMD patients in comparison with age-matched controls, is reduced by GC treatment. Thus, this newly identified pathway may link GC treatment to a mitochondrial response in DMD.
Collapse
Affiliation(s)
- Mathilde Sanson
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Ai Vu Hong
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | | | - Nathalie Bourg
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Laurence Suel
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Fatima Amor
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Guillaume Corre
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Paule Bénit
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Inès Barthélémy
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Stephane Blot
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Anne Bigot
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute, Paris, France
| | | | - Pierre Rustin
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Isabelle Richard
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - David Israeli
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France.
| |
Collapse
|
32
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
33
|
Barthélémy I, Hitte C, Tiret L. The Dog Model in the Spotlight: Legacy of a Trustful Cooperation. J Neuromuscul Dis 2020; 6:421-451. [PMID: 31450509 PMCID: PMC6918919 DOI: 10.3233/jnd-190394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dogs have long been used as a biomedical model system and in particular as a preclinical proof of concept for innovative therapies before translation to humans. A recent example of the utility of this animal model is the promising myotubularin gene delivery in boys affected by X-linked centronuclear myopathy after successful systemic, long-term efficient gene therapy in Labrador retrievers. Mostly, this is due to unique features that make dogs an optimal system. The continuous emergence of spontaneous inherited disorders enables the identification of reliable complementary molecular models for human neuromuscular disorders (NMDs). Dogs’ characteristics including size, lifespan and unprecedented medical care level allow a comprehensive longitudinal description of diseases. Moreover, the highly similar pathogenic mechanisms with human patients yield to translational robustness. Finally, interindividual phenotypic heterogeneity between dogs helps identifying modifiers and anticipates precision medicine issues. This review article summarizes the present list of molecularly characterized dog models for NMDs and provides an exhaustive list of the clinical and paraclinical assays that have been developed. This toolbox offers scientists a sensitive and reliable system to thoroughly evaluate neuromuscular function, as well as efficiency and safety of innovative therapies targeting these NMDs. This review also contextualizes the model by highlighting its unique genetic value, shaped by the long-term coevolution of humans and domesticated dogs. Because the dog is one of the most protected research animal models, there is considerable opposition to include it in preclinical projects, posing a threat to the use of this model. We thus discuss ethical issues, emphasizing that unlike many other models, the dog also benefits from its contribution to comparative biomedical research with a drastic reduction in the prevalence of morbid alleles in the breeding stock and an improvement in medical care.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Christophe Hitte
- CNRS, University of Rennes 1, UMR 6290, IGDR, Faculty of Medicine, SFR Biosit, Rennes, France
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
34
|
van Westering TLE, Lomonosova Y, Coenen-Stass AML, Betts CA, Bhomra A, Hulsker M, Clark LE, McClorey G, Aartsma-Rus A, van Putten M, Wood MJA, Roberts TC. Uniform sarcolemmal dystrophin expression is required to prevent extracellular microRNA release and improve dystrophic pathology. J Cachexia Sarcopenia Muscle 2020; 11:578-593. [PMID: 31849191 PMCID: PMC7113513 DOI: 10.1002/jcsm.12506] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disorder caused by genetic loss of dystrophin protein. Extracellular microRNAs (ex-miRNAs) are putative, minimally invasive biomarkers of DMD. Specific ex-miRNAs (e.g. miR-1, miR-133a, miR-206, and miR-483) are highly up-regulated in the serum of DMD patients and dystrophic animal models and are restored to wild-type levels following exon skipping-mediated dystrophin rescue in mdx mice. As such, ex-miRNAs are promising pharmacodynamic biomarkers of exon skipping efficacy. Here, we aimed to determine the degree to which ex-miRNA levels reflect the underlying level of dystrophin protein expression in dystrophic muscle. METHODS Candidate ex-miRNA biomarker levels were investigated in mdx mice in which dystrophin was restored with peptide-PMO (PPMO) exon skipping conjugates and in mdx-XistΔhs mice that express variable amounts of dystrophin from birth as a consequence of skewed X-chromosome inactivation. miRNA profiling was performed in mdx-XistΔhs mice using the FirePlex methodology and key results validated by small RNA TaqMan RT-qPCR. The muscles from each animal model were further characterized by dystrophin western blot and immunofluorescence staining. RESULTS The restoration of ex-myomiR abundance observed following PPMO treatment was not recapitulated in the high dystrophin-expressing mdx-XistΔhs group, despite these animals expressing similar amounts of total dystrophin protein (~37% of wild-type levels). Instead, ex-miRNAs were present at high levels in mdx-XistΔhs mice regardless of dystrophin expression. PPMO-treated muscles exhibited a uniform pattern of dystrophin localization and were devoid of regenerating fibres, whereas mdx-XistΔhs muscles showed non-homogeneous dystrophin staining and sporadic regenerating foci. CONCLUSIONS Uniform dystrophin expression is required to prevent ex-miRNA release, stabilize myofiber turnover, and attenuate pathology in dystrophic muscle.
Collapse
Affiliation(s)
- Tirsa L E van Westering
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Yulia Lomonosova
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Margriet Hulsker
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lucy E Clark
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK.,Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA, USA
| |
Collapse
|
35
|
O. Mousa N, Osman A, Fahmy N, Abdellatif A, Zada S, El-Fawal H. Duchenne Muscular Dystrophy (DMD) Diagnosis: Past and Present Perspectives. Rare Dis 2020. [DOI: 10.5772/intechopen.90862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
36
|
Mousa NO, Abdellatif A, Fahmy N, Zada S, El-Fawal H, Osman A. Circulating MicroRNAs in Duchenne Muscular Dystrophy. Clin Neurol Neurosurg 2019; 189:105634. [PMID: 31838454 DOI: 10.1016/j.clineuro.2019.105634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/27/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The diagnosis of Duchenne Muscular Dystrophy (DMD) currently depends on non-specific measures such as Creatine kinase (CK) levels. MicroRNAs (miRNAs) are a class of small, endogenous RNAs of 21-25 nucleotides, that are important regulators for numerous physiological and pathological processes. The aim of the current study is to assess the potential of miRNAs as non-invasive biomarker for the diagnosis of DMD and for identifying carriers. PATIENTS AND METHODS Thirty healthy subjects and 29 families with one member diagnosed with DMD were enrolled in the study. Peripheral blood samples were collected from all subjects where microRNAs were extracted from plasma followed by the quantification of miR-499, miR-103a-3p, miR-103a-5p, miR-206, miR-208a, miR-223 and miR-191-5p. MLPA and NGS were carried out as a gold standard technique to identify the mutations in the participants. RESULTS Our data revealed that miR-499 was significantly upregulated in all DMD patients, and true carriers (mothers), while 78 % of potential carriers (sisters) exhibited high levels of this miRNA. Similarly, miR-103a-3p showed an increase in the patients' families although to a lesser extent. On the other hand, miR-206 and miR-191-5p were significantly downregulated in the majority of the DMD patients and the tested female family members. MicroRNA miR-103a-5p and miR-208a followed a comparable trend in patients and mothers. CONCLUSIONS Ourresults suggest that the plasma levels of miRNAs have the capability to diagnose DMD patients and more importantly, miRNAs can be used to identify female carriers.
Collapse
Affiliation(s)
- Nahla O Mousa
- Biology Department, and Biotechnology Program, School of Sciences & Engineering, The American University in Cairo, School of Sciences and Engineering, 11835, Cairo, Egypt; Biotechnology Department, Faculty of Science, Cairo University, 12613, Giza, Egypt
| | - Ahmed Abdellatif
- Biology Department, and Biotechnology Program, School of Sciences & Engineering, The American University in Cairo, School of Sciences and Engineering, 11835, Cairo, Egypt.
| | - Nagia Fahmy
- Neuromuscular Unit, Neuropsychiatry Department, Faculty of Medicine Ain Shams University, 11566, Cairo, Egypt
| | - Suher Zada
- Biology Department, and Biotechnology Program, School of Sciences & Engineering, The American University in Cairo, School of Sciences and Engineering, 11835, Cairo, Egypt
| | - Hassan El-Fawal
- Biology Department, and Biotechnology Program, School of Sciences & Engineering, The American University in Cairo, School of Sciences and Engineering, 11835, Cairo, Egypt
| | - Ahmed Osman
- Biotechnology Department, Basic and Applied Sciences Institute, Egypt-Japan University of Science and Technology, Borg Al Arab, 21934, Egypt; Biochemistry Department, Faculty of Science, Ain Shams University, 11566, Cairo, Egypt.
| |
Collapse
|
37
|
Pegoraro V, Missaglia S, Marozzo R, Tavian D, Angelini C. MiRNAs as biomarkers of phenotype in neutral lipid storage disease with myopathy. Muscle Nerve 2019; 61:253-257. [PMID: 31729045 PMCID: PMC7004093 DOI: 10.1002/mus.26761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neutral lipid storage disease with myopathy (NLSDM) is a rare lipid metabolism disorder. In this study, we evaluated some circulating miRNAs levels in serum samples and the MRI of three affected siblings. METHODS Three members of one NLSDM family were identified: two brothers and one sister. Muscles of lower and right upper extremities were studied by MRI. Expression profile of miRNAs, obtained from serum samples, was detected using qRT-PCR. RESULTS Two brothers presented with progressive skeletal myopathy, while the sister had severe hepatosteatosis and diabetes. NLSDM patients showed a significant increase of muscle-specific miRNAs expression compared with healthy subjects. We found a correlation between hepatic damage and elevation of miRNAs expression profile of liver origin. CONCLUSIONS The dysregulation of miRNAs might represent an indicator of skeletal and hepatic damage and it might be useful to monitor the progression of NLSDM.
Collapse
Affiliation(s)
| | - Sara Missaglia
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | | | - Daniela Tavian
- Laboratory of Cellular Biochemistry and Molecular Biology, CRIBENS, Università Cattolica del Sacro Cuore, Milan, Italy.,Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | | |
Collapse
|
38
|
Kawata R, Yokoi T. Analysis of a Skeletal Muscle Injury and Drug Interactions in Lovastatin- and Fenofibrate-Coadministered Dogs. Int J Toxicol 2019; 38:192-201. [PMID: 31113311 DOI: 10.1177/1091581819844793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Because dogs are widely used in drug development as nonrodent experimental animals, using a dog model for drug-induced adverse reactions is considered to be relevant for an evaluation and investigation of a mechanism and a biomarker of clinical drug-induced adverse reactions. Skeletal muscle injury occurs by various drugs, including statins and fibrates, during drug development. However, there is almost no report of a dog model for drug-induced skeletal muscle injury. In the present study, we induced skeletal muscle injury in dogs by oral coadministration of lovastatin (LV) and fenofibrate (FF) for 4 weeks. Increases in plasma levels of creatine phosphokinase, myoglobin, miR-1, and miR-133a and degeneration/necrosis of myofibers in skeletal muscles but not in the heart were observed in LV- and FF-coadministered dogs. Plasma levels of lovastatin lactone and lovastatin acid were higher in LV- and FF-coadministered dogs than LV-administered dogs. Taken together, FF coadministration is considered to affect LV metabolism and result in skeletal muscle injury.
Collapse
Affiliation(s)
- Reo Kawata
- 1 Division of Clinical Pharmacology, Department of Drug Safety Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsuyoshi Yokoi
- 1 Division of Clinical Pharmacology, Department of Drug Safety Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
39
|
Kostek M. Precision Medicine and Exercise Therapy in Duchenne Muscular Dystrophy. Sports (Basel) 2019; 7:sports7030064. [PMID: 30875955 PMCID: PMC6473733 DOI: 10.3390/sports7030064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 01/15/2023] Open
Abstract
Precision medicine is being discussed and incorporated at all levels of health care and disease prevention, management, and treatment. Key components include new taxonomies of disease classification, the measurement and incorporation of genetics and "omics" data, biomarkers, and health care professionals who can optimize this information for a precision approach to treatment. The study and treatment of Duchenne Muscular Dystrophy is making rapid advances in these areas in addition to rapid advances in new gene and cell-based therapies. New therapies will increase the variability in disease severity, furthering a need for a precision-based approach. An area of therapy that is rarely considered in this approach is how the physiology of muscle contractions will interact with these therapies and a precision approach. As muscle pathology improves, physical activity levels will increase, which will likely be very beneficial to some patients but likely not to all. Physical activity is likely to synergistically improve these therapies and can be used to enhance muscle health and quality of life after these therapies are delivered using the tools of precision medicine.
Collapse
Affiliation(s)
- Matthew Kostek
- Laboratory of Muscle and Translational Therapeutics, Department of Physical Therapy, Duquesne University, Pittsburgh, PA 15228, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15228, USA.
| |
Collapse
|
40
|
Shibasaki H, Imamura M, Arima S, Tanihata J, Kuraoka M, Matsuzaka Y, Uchiumi F, Tanuma SI, Takeda S. Characterization of a novel microRNA, miR-188, elevated in serum of muscular dystrophy dog model. PLoS One 2019; 14:e0211597. [PMID: 30699200 PMCID: PMC6353185 DOI: 10.1371/journal.pone.0211597] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/16/2019] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding small RNAs that regulate gene expression at the post-transcriptional level. Several miRNAs are exclusively expressed in skeletal muscle and participate in the regulation of muscle differentiation by interacting with myogenic factors. These miRNAs can be found at high levels in the serum of patients and animal models for Duchenne muscular dystrophy, which is expected to be useful as biomarkers for their clinical conditions. By miRNA microarray analysis, we identified miR-188 as a novel miRNA that is elevated in the serum of the muscular dystrophy dog model, CXMDJ. miR-188 was not muscle-specific miRNA, but its expression was up-regulated in skeletal muscles associated with muscle regeneration induced by cardiotoxin-injection in normal dogs and mice. Manipulation of miR-188 expression using antisense oligo and mimic oligo RNAs alters the mRNA expression of the myogenic regulatory factors, MRF4 and MEF2C. Our results suggest that miR-188 is a new player that participates in the gene regulation process of muscle differentiation and that it may serve as a serum biomarker reflecting skeletal muscle regeneration.
Collapse
Affiliation(s)
- Hiroyuki Shibasaki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Michihiro Imamura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Sayuri Arima
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of Cell Physiology, The Jikei University School of Medicine, Minato, Tokyo, Japan
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Laboratory of Experimental Animal Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Yasunari Matsuzaka
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Fumiaki Uchiumi
- Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Sei-Ichi Tanuma
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
41
|
Kasimanickam V, Buhr M, Kasimanickam R. Patterns of expression of sperm and seminal plasma microRNAs in boar semen. Theriogenology 2018; 125:87-92. [PMID: 30391831 DOI: 10.1016/j.theriogenology.2018.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/26/2018] [Indexed: 01/15/2023]
Abstract
Although sperm and seminal plasma differ in their origin, biophysical and biochemical properties of seminal plasma influence the sperm function. Seminal plasma is a fluid medium containing substances from testes, epididymides and accessory glands. Composition of seminal plasma varies among animal species and in boars, prostate and bulbourethral glands are major contributors to the volume and contents. While the origin of some components of seminal plasma are known, the source of recently discovered seminal plasma microRNAs remains unknown, in part due to the difficulty of recovering and characterizing RNA from porcine sperm and seminal plasma. To test the hypothesis that seminal plasma miRNAs interact with sperm, the first objective was to validate protocols for recovering RNAs from porcine seminal plasma and sperm, whereas the second objective was to characterize expression patterns of 84 prioritized microRNAs employing real time PCR methodology. The study identified a relationship between sperm and seminal plasma microRNAs, based on the normalized threshold cycle of amplifying cDNA in sperm and seminal plasma from the same semen of Landrace boars. Therefore, it was concluded that seminal plasma miRNAs may originate from sperm or these miRNAs may shuttle between sperm and seminal plasma in order to facilitate cell-to-cell communication.
Collapse
Affiliation(s)
- Vanmathy Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA; Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| | - Mary Buhr
- College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK, S7N 5A8, Canada
| | - Ramanathan Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
42
|
Spitali P, Hettne K, Tsonaka R, Charrout M, van den Bergen J, Koeks Z, Kan HE, Hooijmans MT, Roos A, Straub V, Muntoni F, Al-Khalili-Szigyarto C, Koel-Simmelink MJA, Teunissen CE, Lochmüller H, Niks EH, Aartsma-Rus A. Tracking disease progression non-invasively in Duchenne and Becker muscular dystrophies. J Cachexia Sarcopenia Muscle 2018; 9:715-726. [PMID: 29682908 PMCID: PMC6104105 DOI: 10.1002/jcsm.12304] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/25/2018] [Accepted: 03/10/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Analysis of muscle biopsies allowed to characterize the pathophysiological changes of Duchenne and Becker muscular dystrophies (D/BMD) leading to the clinical phenotype. Muscle tissue is often investigated during interventional dose finding studies to show in situ proof of concept and pharmacodynamics effect of the tested drug. Less invasive readouts are needed to objectively monitor patients' health status, muscle quality, and response to treatment. The identification of serum biomarkers correlating with clinical function and able to anticipate functional scales is particularly needed for personalized patient management and to support drug development programs. METHODS A large-scale proteomic approach was used to identify serum biomarkers describing pathophysiological changes (e.g. loss of muscle mass), association with clinical function, prediction of disease milestones, association with in vivo 31 P magnetic resonance spectroscopy data and dystrophin levels in muscles. Cross-sectional comparisons were performed to compare DMD patients, BMD patients, and healthy controls. A group of DMD patients was followed up for a median of 4.4 years to allow monitoring of individual disease trajectories based on yearly visits. RESULTS Cross-sectional comparison enabled to identify 10 proteins discriminating between healthy controls, DMD and BMD patients. Several proteins (285) were able to separate DMD from healthy, while 121 proteins differentiated between BMD and DMD; only 13 proteins separated BMD and healthy individuals. The concentration of specific proteins in serum was significantly associated with patients' performance (e.g. BMP6 serum levels and elbow flexion) or dystrophin levels (e.g. TIMP2) in BMD patients. Analysis of longitudinal trajectories allowed to identify 427 proteins affected over time indicating loss of muscle mass, replacement of muscle by adipose tissue, and cardiac involvement. Over-representation analysis of longitudinal data allowed to highlight proteins that could be used as pharmacodynamic biomarkers for drugs currently in clinical development. CONCLUSIONS Serum proteomic analysis allowed to not only discriminate among DMD, BMD, and healthy subjects, but it enabled to detect significant associations with clinical function, dystrophin levels, and disease progression.
Collapse
Affiliation(s)
- Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kristina Hettne
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Roula Tsonaka
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mohammed Charrout
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Zaïda Koeks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermien E Kan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa T Hooijmans
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Roos
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, University of Newcastle, Newcastle upon Tyne, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, University of Newcastle, Newcastle upon Tyne, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health, London, UK
| | | | - Marleen J A Koel-Simmelink
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, The Netherlands
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, University of Newcastle, Newcastle upon Tyne, UK
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
Nicking-enhanced rolling circle amplification for sensitive fluorescent detection of cancer-related microRNAs. Anal Bioanal Chem 2018; 410:6819-6826. [PMID: 30066196 DOI: 10.1007/s00216-018-1277-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 01/23/2023]
Abstract
In this study, a biosensing system based on nicking-enhanced rolling circle amplification (N-RCA) was proposed for the highly sensitive detection of cancer-related let-7a microRNA (miRNA). The sensing system consists of a padlock probe (PP), which contains a target recognition sequence and two binding sites for nicking endonuclease (NEase), and molecular beacon (MB) as reporting molecule. Upon hybridization with let-7a, the PP can be circularized by ligase. Then, the miRNA acted as polymerization primer to initiate rolling circle amplification (RCA). With the assistance of NEase, RCA products can be nicked on the cyclized PP and are displaced during the subsequent duplication process, generating numerous nicked fragments (NFs). These NFs not only induce another RCA reaction but also open the molecular beacons (MBs) via hybridization, leading to significantly amplified fluorescence signal. Under the optimized conditions, this method exhibits high sensitivity toward target miRNA let-7a with a detection limit of as low as 10 pM, a dynamic range of three orders of magnitude is achieved, and its family member is easily distinguished even with only one mismatched base. Meanwhile, it displays good recovery and satisfactory reproducibility in fetal bovine serum (FBS). Therefore, these merits endow the newly proposed N-RCA strategy with powerful implications for miRNA detection. Graphical abstract A biosensing system based on nicking-enhanced rolling circle amplification (N-RCA) for the highly sensitive detection of cancer-related let-7a microRNA.
Collapse
|
44
|
Siracusa J, Koulmann N, Sourdrille A, Chapus C, Verret C, Bourdon S, Goriot ME, Banzet S. Phenotype-Specific Response of Circulating miRNAs Provides New Biomarkers of Slow or Fast Muscle Damage. Front Physiol 2018; 9:684. [PMID: 29922177 PMCID: PMC5996145 DOI: 10.3389/fphys.2018.00684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/16/2018] [Indexed: 01/19/2023] Open
Abstract
Skeletal muscle is a heterogeneous tissue composed of a continuum of contracting fibers ranging from slow-type to fast-type fibers. Muscle damage is a frequent event and a susceptibility of fast-fibers to exercise-induced damage (EIMD) or statins toxicity has been reported. Biological markers of muscle damage such as creatine kinase (CK) are not fiber-type specific and new biomarkers are needed. Some microRNAs (miRNAs) are specific to the muscle tissue, can be found in the extracellular compartment and can rise in the plasma following muscle damage. Our aim was to identify whether a set of circulating miRNAs can be used as fiber-type specific biomarkers of muscle damage in a model of traumatic (crush) injuries induced either in the slow soleus (SOL) or in the fast extensor digitorum longus (EDL) muscles of rats. A subset of miRNAs composed of miR-1-3p, -133a-3p, -133b-3p, 206-3p, -208b-3p, 378a-3p, -434-3p, and -499-5p were measured by RT-PCR in non-injured SOL or EDL muscle and in the plasma of rats 12 h after damage induced to SOL or EDL. MiR-133b-3p, -378a-3p, and -434-3p were equally expressed both in SOL and EDL muscles. MiR-1-3-p and -133a-3p levels were higher in EDL compared to SOL (1.3- and 1.1-fold, respectively). Conversely, miR-206-3p, -208b-3p, and -499-5p were mainly expressed in SOL compared to EDL (7.4-, 35.4-, and 10.7-fold, respectively). In the plasma, miR-1-3p and -133a-3p were elevated following muscle damage compared to a control group, with no difference between SOL and EDL. MiR-133b-3p and -434-3p plasma levels were significantly higher in EDL compared to SOL (1.8- and 2.4-fold, respectively), while miR-378a-3p rose only in the EDL group. MiR-206-3p levels were elevated in SOL only (fourfold compared to EDL). Our results show that plasma miR-133b-3p and -434 are fast-fiber specific biomarkers, while miR-206-3p is a robust indicator of slow-fiber damage, opening new perspectives to monitor fiber-type selective muscle damage in research and clinic.
Collapse
Affiliation(s)
- Julien Siracusa
- Unité de Physiologie de l'Exercice et des Activités en Conditions Extrêmes, Département Environnements Opérationnels, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Nathalie Koulmann
- Unité de Physiologie de l'Exercice et des Activités en Conditions Extrêmes, Département Environnements Opérationnels, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France.,Ecole du Val-de-Grâce, Paris, France
| | - Antoine Sourdrille
- Unité de Physiologie de l'Exercice et des Activités en Conditions Extrêmes, Département Environnements Opérationnels, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Charles Chapus
- Unité de Biologie Moléculaire, Département des Plateformes et Recherche Technologique, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Catherine Verret
- Bureau de Gestion de Recherche Clinique, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Stéphanie Bourdon
- Unité de Physiologie de l'Exercice et des Activités en Conditions Extrêmes, Département Environnements Opérationnels, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Marie-Emmanuelle Goriot
- Unité de Thérapie Tissulaire et Traumatologie de Guerre, Département Soutien Médico-Chirurgical des Forces, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France.,INSERM U 1197, Clamart, France
| | - Sébastien Banzet
- Ecole du Val-de-Grâce, Paris, France.,Unité de Thérapie Tissulaire et Traumatologie de Guerre, Département Soutien Médico-Chirurgical des Forces, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France.,INSERM U 1197, Clamart, France
| |
Collapse
|
45
|
Guilbaud M, Gentil C, Peccate C, Gargaun E, Holtzmann I, Gruszczynski C, Falcone S, Mamchaoui K, Ben Yaou R, Leturcq F, Jeanson-Leh L, Piétri-Rouxel F. miR-708-5p and miR-34c-5p are involved in nNOS regulation in dystrophic context. Skelet Muscle 2018; 8:15. [PMID: 29703249 PMCID: PMC5924477 DOI: 10.1186/s13395-018-0161-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/30/2022] Open
Abstract
Background Duchenne (DMD) and Becker (BMD) muscular dystrophies are caused by mutations in the DMD gene coding for dystrophin, a protein being part of a large sarcolemmal protein scaffold that includes the neuronal nitric oxide synthase (nNOS). The nNOS was shown to play critical roles in a variety of muscle functions and alterations of its expression and location in dystrophic muscle fiber leads to an increase of the muscle fatigability. We previously revealed a decrease of nNOS expression in BMD patients all presenting a deletion of exons 45 to 55 in the DMD gene (BMDd45-55), impacting the nNOS binding site of dystrophin. Since several studies showed deregulation of microRNAs (miRNAs) in dystrophinopathies, we focused on miRNAs that could target nNOS in dystrophic context. Methods By a screening of 617 miRNAs in BMDd45-55 muscular biopsies using TLDA and an in silico study to determine which one could target nNOS, we selected four miRNAs. In order to select those that targeted a sequence of 3′UTR of NOS1, we performed luciferase gene reporter assay in HEK393T cells. Finally, expression of candidate miRNAs was modulated in control and DMD human myoblasts (DMDd45-52) to study their ability to target nNOS. Results TLDA assay and the in silico study allowed us to select four miRNAs overexpressed in muscle biopsies of BMDd45-55 compared to controls. Among them, only the overexpression of miR-31, miR-708, and miR-34c led to a decrease of luciferase activity in an NOS1-3′UTR-luciferase assay, confirming their interaction with the NOS1-3′UTR. The effect of these three miRNAs was investigated on control and DMDd45-52 myoblasts. First, we showed a decrease of nNOS expression when miR-708 or miR-34c were overexpressed in control myoblasts. We then confirmed that DMDd45-52 cells displayed an endogenous increased of miR-31, miR-708, and miR-34c and a decreased of nNOS expression, the same characteristics observed in BMDd45-55 biopsies. In DMDd45-52 cells, we demonstrated that the inhibition of miR-708 and miR-34c increased nNOS expression, confirming that both miRNAs can modulate nNOS expression in human myoblasts. Conclusion These results strongly suggest that miR-708 and miR-34c, overexpressed in dystrophic context, are new actors involved in the regulation of nNOS expression in dystrophic muscle. Electronic supplementary material The online version of this article (10.1186/s13395-018-0161-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marine Guilbaud
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Christel Gentil
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Cécile Peccate
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Elena Gargaun
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Isabelle Holtzmann
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Carole Gruszczynski
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Sestina Falcone
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Kamel Mamchaoui
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France
| | - Rabah Ben Yaou
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.,AP-HP, Centre de Référence Maladies Neuromusculaire Nord, Est, Ile-de-France, G.H. Pitié-Salpêtrière, F-75013, Paris, France
| | - France Leturcq
- Laboratoire de Génétique et Biologie Moléculaire, Hôpital Cochin, Paris, France
| | | | - France Piétri-Rouxel
- Sorbonne Université-UMRS974-Inserm-Institut de Myologie, 105 bd de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
46
|
Catapano F, Domingos J, Perry M, Ricotti V, Phillips L, Servais L, Seferian A, Groot ID, Krom YD, Niks EH, Verschuuren JJ, Straub V, Voit T, Morgan J, Muntoni F. Downregulation of miRNA-29, -23 and -21 in urine of Duchenne muscular dystrophy patients. Epigenomics 2018; 10:875-889. [PMID: 29564913 DOI: 10.2217/epi-2018-0022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM To study the signature of 87 urinary miRNAs in Duchenne muscular dystrophy (DMD) patients, select the most dysregulated and determine statistically significant differences in their expression between controls, ambulant (A) and nonambulant (NA) DMD patients, and patients on different corticosteroid regimens. Patients/materials & methods: Urine was collected from control (n = 20), A (n = 31) and NA (n = 23) DMD patients. miRNA expression was measured by reverse transcription-quantitative PCR. RESULTS miR-29c-3p was significantly downregulated in A DMD patients while miR-23b-3p and miR-21-5p were significantly downregulated in NA DMD patients compared with age-matched controls. CONCLUSION miR-29c-3p, miR-23b-3p and miR-21-5p are promising novel noninvasive biomarkers for DMD, and miR-29c-3p levels are differentially affected by different steroid regimens, supporting the antifibrotic effect of steroid therapy.
Collapse
Affiliation(s)
- Francesco Catapano
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | - Joana Domingos
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | - Mark Perry
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, St Michael's Building, Portsmouth PO1 2DT, UK
| | - Valeria Ricotti
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | - Lauren Phillips
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Laurent Servais
- Institute I-Motion, Hôpital Armand Trousseau, Paris 75571-12, France.,Centre de Référence des maladies Neuromusculaires, CHU de Liège, Liège 4000, Belgium
| | - Andreea Seferian
- Institute I-Motion, Hôpital Armand Trousseau, Paris 75571-12, France
| | - Imelda de Groot
- Department of Rehabilitation, Amalia Children's Hospital, Radboud University Medical Centre, Nijmegen 6525 GA, The Netherlands
| | - Yvonne D Krom
- Department of Neurology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Jan Jgm Verschuuren
- Department of Neurology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Volker Straub
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Human Genetics, International Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Thomas Voit
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Jennifer Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| |
Collapse
|
47
|
Filipova D, Henry M, Rotshteyn T, Brunn A, Carstov M, Deckert M, Hescheler J, Sachinidis A, Pfitzer G, Papadopoulos S. Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Cav1.1 converge at E18.5. PLoS One 2018; 13:e0194428. [PMID: 29543863 PMCID: PMC5854361 DOI: 10.1371/journal.pone.0194428] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/04/2018] [Indexed: 12/20/2022] Open
Abstract
In skeletal muscle the coordinated actions of two mechanically coupled Ca2+ channels-the 1,4-dihydropyridine receptor (Cav1.1) and the type 1 ryanodine receptor (RYR1)-underlie the molecular mechanism of rapid cytosolic [Ca2+] increase leading to contraction. While both [Ca2+]i and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Cav1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 -the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 -the end of secondary myogenesis, in WT, RYR1-/-, and Cav1.1-/- murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Cav1.1-/- muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1-/- and Cav1.1-/- samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1-/-, and lipid metabolism in Cav1.1-/- samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1-/- and Cav1.1-/- mice. Our results suggest distinct functional roles for RYR1 and Cav1.1 in skeletal primary and secondary myogenesis.
Collapse
Affiliation(s)
- Dilyana Filipova
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Margit Henry
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Tamara Rotshteyn
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Brunn
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Mariana Carstov
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Martina Deckert
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
48
|
Spitali P, Hettne K, Tsonaka R, Sabir E, Seyer A, Hemerik JBA, Goeman JJ, Picillo E, Ergoli M, Politano L, Aartsma-Rus A. Cross-sectional serum metabolomic study of multiple forms of muscular dystrophy. J Cell Mol Med 2018; 22:2442-2448. [PMID: 29441734 PMCID: PMC5867073 DOI: 10.1111/jcmm.13543] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/13/2017] [Indexed: 12/25/2022] Open
Abstract
Muscular dystrophies are characterized by a progressive loss of muscle tissue and/or muscle function. While metabolic alterations have been described in patients’‐derived muscle biopsies, non‐invasive readouts able to describe these alterations are needed in order to objectively monitor muscle condition and response to treatment targeting metabolic abnormalities. We used a metabolomic approach to study metabolites concentration in serum of patients affected by multiple forms of muscular dystrophy such as Duchenne and Becker muscular dystrophies, limb‐girdle muscular dystrophies type 2A and 2B, myotonic dystrophy type 1 and facioscapulohumeral muscular dystrophy. We show that 15 metabolites involved in energy production, amino acid metabolism, testosterone metabolism and response to treatment with glucocorticoids were differentially expressed between healthy controls and Duchenne patients. Five metabolites were also able to discriminate other forms of muscular dystrophy. In particular, creatinine and the creatine/creatinine ratio were significantly associated with Duchenne patients performance as assessed by the 6‐minute walk test and north star ambulatory assessment. The obtained results provide evidence that metabolomics analysis of serum samples can provide useful information regarding muscle condition and response to treatment, such as to glucocorticoids treatment.
Collapse
Affiliation(s)
- Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kristina Hettne
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Roula Tsonaka
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ekrem Sabir
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jesse B A Hemerik
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jelle J Goeman
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Picillo
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Manuela Ergoli
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Siracusa J, Koulmann N, Banzet S. Circulating myomiRs: a new class of biomarkers to monitor skeletal muscle in physiology and medicine. J Cachexia Sarcopenia Muscle 2018; 9:20-27. [PMID: 29193905 PMCID: PMC5803618 DOI: 10.1002/jcsm.12227] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/13/2017] [Accepted: 07/05/2017] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNA) are small non-coding RNAs that target mRNAs and are consequently involved in the post-transcriptional regulation of gene expression. Some miRNAs are ubiquitously expressed in tissue, while others are tissue-specific or tissue-enriched. miRNAs can be released by cells and are found in various biofluids, including serum and plasma. Thus, measuring miRNAs in the circulation may provide information on the originating tissue or cells. MyomiRs are described as striated muscle-specific or muscle-enriched miRNAs. Their circulating levels can be measured and have been proposed to be new biomarkers of physiological and pathological muscle processes. The aims of this review are to summarize the current knowledge of circulating myomiRs, to identify the types of information they can provide about skeletal muscle, and to determine how to apply that information in the fields of research and medicine.
Collapse
Affiliation(s)
- Julien Siracusa
- Institut de Recherche Biomédicale des Armées, 1 place Valérie André, BP73, 91220, Brétigny sur Orge, France
| | - Nathalie Koulmann
- Institut de Recherche Biomédicale des Armées, 1 place Valérie André, BP73, 91220, Brétigny sur Orge, France.,Ecole du Val de Grâce, 1 place Alphonse Laveran, 75005, Paris, France
| | - Sébastien Banzet
- Ecole du Val de Grâce, 1 place Alphonse Laveran, 75005, Paris, France.,Institut de Recherche Biomédicale des Armées, 1 Rue Lieutenant Raoul Batany, 92140, Clamart, France.,INSERM UMRS1197, 1 Rue Lieutenant Raoul Batany, 92140, Clamart, France
| |
Collapse
|
50
|
Szigyarto CAK, Spitali P. Biomarkers of Duchenne muscular dystrophy: current findings. Degener Neurol Neuromuscul Dis 2018; 8:1-13. [PMID: 30050384 PMCID: PMC6053903 DOI: 10.2147/dnnd.s121099] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Numerous biomarkers have been unveiled in the rapidly evolving biomarker discovery field, with an aim to improve the clinical management of disorders. In rare diseases, such as Duchenne muscular dystrophy, this endeavor has created a wealth of knowledge that, if effectively exploited, will benefit affected individuals, with respect to health care, therapy, improved quality of life and increased life expectancy. The most promising findings and molecular biomarkers are inspected in this review, with an aim to provide an overview of currently known biomarkers and the technological developments used. Biomarkers as cells, genetic variations, miRNAs, proteins, lipids and/or metabolites indicative of disease severity, progression and treatment response have the potential to improve development and approval of therapies, clinical management of DMD and patients’ life quality. We highlight the complexity of translating research results to clinical use, emphasizing the need for biomarkers, fit for purpose and describe the challenges associated with qualifying biomarkers for clinical applications.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Division of Proteomics, School of Biotechnology, AlbaNova University Center, KTH-Royal Institute of Technology, Stockholm, Sweden, .,Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden,
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands,
| |
Collapse
|