51
|
226 th ENMC International Workshop:: Towards validated and qualified biomarkers for therapy development for Duchenne muscular dystrophy 20-22 January 2017, Heemskerk, The Netherlands. Neuromuscul Disord 2017; 28:77-86. [PMID: 29203356 DOI: 10.1016/j.nmd.2017.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 12/14/2022]
|
52
|
Coenen-Stass AML, Wood MJA, Roberts TC. Biomarker Potential of Extracellular miRNAs in Duchenne Muscular Dystrophy. Trends Mol Med 2017; 23:989-1001. [PMID: 28988850 DOI: 10.1016/j.molmed.2017.09.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022]
Abstract
miRNAs are small, noncoding RNAs that not only regulate gene expression within cells, but might also constitute promising extracellular biomarkers for a variety of pathologies, including the progressive muscle-wasting disorder Duchenne Muscular Dystrophy (DMD). A set of muscle-enriched miRNAs, the myomiRs (miR-1, miR-133, and miR-206) are highly elevated in the serum of patients with DMD and in dystrophin-deficient animal models. Furthermore, circulating myomiRs might be used as pharmacodynamic biomarkers, given that their levels can be restored towards wild-type levels following exon skipping therapy in dystrophic mice. The relationship between muscle pathology and extracellular myomiR release is complex, and incompletely understood. Here, we discuss current progress leading towards the clinical utility of extracellular miRNAs as putative DMD biomarkers, and their possible contribution to muscle physiology.
Collapse
Affiliation(s)
- Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; Institute of Neurology, Sobell Department of Motor Neuroscience and Movement Disorders, University College London, London, Queen Square, London, WC1N 3BG, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA 92037, USA.
| |
Collapse
|
53
|
Marsolier J, Laforet P, Pegoraro E, Vissing J, Richard I. 1st International Workshop on Clinical trial readiness for sarcoglycanopathies 15-16 November 2016, Evry, France. Neuromuscul Disord 2017; 27:683-692. [PMID: 28521973 DOI: 10.1016/j.nmd.2017.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 02/14/2017] [Indexed: 11/25/2022]
Affiliation(s)
- Justine Marsolier
- Généthon, INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | | | | | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Isabelle Richard
- Généthon, INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France.
| | | |
Collapse
|
54
|
Amirouche A, Jahnke VE, Lunde JA, Koulmann N, Freyssenet DG, Jasmin BJ. Muscle-specific microRNA-206 targets multiple components in dystrophic skeletal muscle representing beneficial adaptations. Am J Physiol Cell Physiol 2017; 312:C209-C221. [DOI: 10.1152/ajpcell.00185.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/06/2016] [Accepted: 12/21/2016] [Indexed: 01/31/2023]
Abstract
Over the last several years, converging lines of evidence have indicated that miR-206 plays a pivotal role in promoting muscle differentiation and regeneration, thereby potentially impacting positively on the progression of neuromuscular disorders, including Duchenne muscular dystrophy (DMD). Despite several studies showing the regulatory function of miR-206 on target mRNAs in skeletal muscle cells, the effects of overexpression of miR-206 in dystrophic muscles remain to be established. Here, we found that miR-206 overexpression in mdx mouse muscles simultaneously targets multiple mRNAs and proteins implicated in satellite cell differentiation, muscle regeneration, and at the neuromuscular junction. Overexpression of miR-206 also increased the levels of several muscle-specific mRNAs/proteins, while enhancing utrophin A expression at the sarcolemma. Finally, we also observed that the increased expression of miR-206 in dystrophin-deficient mouse muscle decreased the production of proinflammatory cytokines and infiltration of macrophages. Taken together, our results show that miR-206 acts as a pleiotropic regulator that targets multiple key mRNAs and proteins expected to provide beneficial adaptations in dystrophic muscle, thus highlighting its therapeutic potential for DMD.
Collapse
Affiliation(s)
- Adel Amirouche
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Vanessa E. Jahnke
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - John A. Lunde
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Nathalie Koulmann
- Institut de Recherche Biomédicale des Armées, Département Environnements Opérationnels, Bretigny-sur-Orge, France
| | - Damien G. Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université de Saint Etienne, Université de Lyon, Lyon, France
| | - Bernard J. Jasmin
- Department of Cellular and Molecular Medicine and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| |
Collapse
|
55
|
Liu J, Liang X, Zhou D, Lai L, Xiao L, Liu L, Fu T, Kong Y, Zhou Q, Vega RB, Zhu MS, Kelly DP, Gao X, Gan Z. Coupling of mitochondrial function and skeletal muscle fiber type by a miR-499/Fnip1/AMPK circuit. EMBO Mol Med 2016; 8:1212-1228. [PMID: 27506764 PMCID: PMC5048369 DOI: 10.15252/emmm.201606372] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Upon adaption of skeletal muscle to physiological and pathophysiological stimuli, muscle fiber type and mitochondrial function are coordinately regulated. Recent studies have identified pathways involved in control of contractile proteins of oxidative-type fibers. However, the mechanism for coupling of mitochondrial function to the muscle contractile machinery during fiber type transition remains unknown. Here, we show that the expression of the genes encoding type I myosins, Myh7/Myh7b and their intronic miR-208b/miR-499, parallels mitochondrial function during fiber type transitions. Using in vivo approaches in mice, we found that miR-499 drives a PGC-1α-dependent mitochondrial oxidative metabolism program to match shifts in slow-twitch muscle fiber composition. Mechanistically, miR-499 directly targets Fnip1, an AMP-activated protein kinase (AMPK)-interacting protein that negatively regulates AMPK, a known activator of PGC-1α. Inhibition of Fnip1 reactivated AMPK/PGC-1α signaling and mitochondrial function in myocytes. Restoration of the expression of miR-499 in the mdx mouse model of Duchenne muscular dystrophy (DMD) reduced the severity of DMD Thus, we have identified a miR-499/Fnip1/AMPK circuit that can serve as a mechanism to couple muscle fiber type and mitochondrial function.
Collapse
Affiliation(s)
- Jing Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xijun Liang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Danxia Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Ling Lai
- Diabetes and Obesity Research Center, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, USA
| | - Liwei Xiao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Lin Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Tingting Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yan Kong
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
| | - Qian Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Rick B Vega
- Diabetes and Obesity Research Center, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, USA
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Daniel P Kelly
- Diabetes and Obesity Research Center, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, USA
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| |
Collapse
|
56
|
Abstract
Noncoding RNAs (ncRNAs) such as miRNAs and long noncoding RNAs modulate gene transcription in response to environmental stressors and other stimuli. A role for ncRNAs in muscle pathologies has been demonstrated and further evidence suggests that ncRNAs also play a role in Duchenne muscular dystrophy (DMD). Studies investigating the differential expression of miRNAs in biological fluids between DMD patients and models of dystrophin deficiency (the MDX mouse model, canine models of DMD) and controls have been published, as these have a role in fibrosis. Long noncoding RNAs are differentially expressed in DMD patients and may, in part, have a mechanism of action via targeting of miRNAs. Although many of these recent findings need to be confirmed, ncRNAs may prove to be useful as potential biomarkers of disease. However, their use as therapeutic targets in DMD remains unclear.
Collapse
Affiliation(s)
- Mark M Perry
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, UK
| |
Collapse
|
57
|
Arvanitidis A, Henriksen K, Karsdal M, Nedergaard A. Neo-epitope Peptides as Biomarkers of Disease Progression for Muscular Dystrophies and Other Myopathies. J Neuromuscul Dis 2016; 3:333-346. [PMID: 27854226 PMCID: PMC5123625 DOI: 10.3233/jnd-160150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
For several decades, serological biomarkers of neuromuscular diseases as dystrophies, myopathies and myositis have been limited to routine clinical biochemistry panels. Gauging the pathological progression is a prerequisite for proper treatment and therefore identifying accessible, easy to monitor biomarkers that can predict the disease progression would be an important advancement. Most muscle diseases involve accelerated muscle fiber degradation, inflammation, fatty tissue substitution and/or fibrosis. All these pathological traits have been shown to give rise to serological peptide biomarkers in other tissues, underlining the potential application of existing biomarkers of such traits in muscle disorders. A significant quantity of tissue is involved in these pathological mechanisms alongside with qualitative changes in protein turnover in myofibrillar, extra-cellular matrix and immunological cell protein fractions accompanied by alterations in body fluids. We propose that protein and peptides can leak out of the afflicted muscles and can be of use in diagnosis, prediction of pathology trajectory and treatment efficacy. Proteolytic cleavage systems are especially modulated during a range of muscle pathologies, thereby giving rise to peptides that are differentially released during disease manifestation. Therefore, we believe that pathology-specific post-translational modifications like cleavages can give rise to neoepitope peptides that may represent a promising class of peptides for discovery of biomarkers pertaining to neuromuscular diseases.
Collapse
Affiliation(s)
- A. Arvanitidis
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - K. Henriksen
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - M.A. Karsdal
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| | - A. Nedergaard
- Nordic Bioscience, Musculoskeletal Diseases, Herlev, Denmark
| |
Collapse
|
58
|
Circulating miRNAs are generic and versatile therapeutic monitoring biomarkers in muscular dystrophies. Sci Rep 2016; 6:28097. [PMID: 27323895 PMCID: PMC4914855 DOI: 10.1038/srep28097] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/26/2016] [Indexed: 12/29/2022] Open
Abstract
The development of medical approaches requires preclinical and clinical trials for assessment of therapeutic efficacy. Such evaluation entails the use of biomarkers, which provide information on the response to the therapeutic intervention. One newly-proposed class of biomarkers is the microRNA (miRNA) molecules. In muscular dystrophies (MD), the dysregulation of miRNAs was initially observed in muscle biopsy and later extended to plasma samples, suggesting that they may be of interest as biomarkers. First, we demonstrated that dystromiRs dysregulation occurs in MD with either preserved or disrupted expression of the dystrophin-associated glycoprotein complex, supporting the utilization of dystromiRs as generic biomarkers in MD. Then, we aimed at evaluation of the capacity of miRNAs as monitoring biomarkers for experimental therapeutic approach in MD. To this end, we took advantage of our previously characterized gene therapy approach in a mouse model for α-sarcoglycanopathy. We identified a dose-response correlation between the expression of miRNAs on both muscle tissue and blood serum and the therapeutic benefit as evaluated by a set of new and classically-used evaluation methods. This study supports the utility of profiling circulating miRNAs for the evaluation of therapeutic outcome in medical approaches for MD.
Collapse
|
59
|
Nie M, Liu J, Yang Q, Seok HY, Hu X, Deng ZL, Wang DZ. MicroRNA-155 facilitates skeletal muscle regeneration by balancing pro- and anti-inflammatory macrophages. Cell Death Dis 2016; 7:e2261. [PMID: 27277683 PMCID: PMC5143393 DOI: 10.1038/cddis.2016.165] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/26/2016] [Accepted: 05/12/2016] [Indexed: 12/19/2022]
Abstract
Skeletal muscle has remarkable regeneration capacity and regenerates in response to injury. Muscle regeneration largely relies on muscle stem cells called satellite cells. Satellite cells normally remain quiescent, but in response to injury or exercise they become activated and proliferate, migrate, differentiate, and fuse to form multinucleate myofibers. Interestingly, the inflammatory process following injury and the activation of the myogenic program are highly coordinated, with myeloid cells having a central role in modulating satellite cell activation and regeneration. Here, we show that genetic deletion of microRNA-155 (miR-155) in mice substantially delays muscle regeneration. Surprisingly, miR-155 does not appear to directly regulate the proliferation or differentiation of satellite cells. Instead, miR-155 is highly expressed in myeloid cells, is essential for appropriate activation of myeloid cells, and regulates the balance between pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages during skeletal muscle regeneration. Mechanistically, we found that miR-155 suppresses SOCS1, a negative regulator of the JAK-STAT signaling pathway, during the initial inflammatory response upon muscle injury. Our findings thus reveal a novel role of miR-155 in regulating initial immune responses during muscle regeneration and provide a novel miRNA target for improving muscle regeneration in degenerative muscle diseases.
Collapse
Affiliation(s)
- M Nie
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, P.R. China
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
| | - J Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
| | - Q Yang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, P.R. China
| | - H Y Seok
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
| | - X Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
| | - Z-L Deng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, P.R. China
| | - D-Z Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA, USA
| |
Collapse
|
60
|
Ballarino M, Morlando M, Fatica A, Bozzoni I. Non-coding RNAs in muscle differentiation and musculoskeletal disease. J Clin Invest 2016; 126:2021-30. [PMID: 27249675 DOI: 10.1172/jci84419] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RNA is likely to be the most rediscovered macromolecule in biology. Periodically, new non-canonical functions have been ascribed to RNA, such as the ability to act as a catalytic molecule or to work independently from its coding capacity. Recent annotations show that more than half of the transcriptome encodes for RNA molecules lacking coding activity. Here we illustrate how these transcripts affect skeletal muscle differentiation and related disorders. We discuss the most recent scientific discoveries that have led to the identification of the molecular circuitries that are controlled by RNA during the differentiation process and that, when deregulated, lead to pathogenic events. These findings will provide insights that can aid in the development of new therapeutic interventions for muscle diseases.
Collapse
MESH Headings
- Animals
- Biomarkers/blood
- Cell Differentiation
- Genetic Markers
- Humans
- Mice
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Models, Biological
- Muscle Development/genetics
- Muscle Development/physiology
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- Musculoskeletal Diseases/genetics
- Musculoskeletal Diseases/metabolism
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Untranslated/blood
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Transcriptome
Collapse
|
61
|
Robriquet F, Babarit C, Larcher T, Dubreil L, Ledevin M, Goubin H, Rouger K, Guével L. Identification in GRMD dog muscle of critical miRNAs involved in pathophysiology and effects associated with MuStem cell transplantation. BMC Musculoskelet Disord 2016; 17:209. [PMID: 27170302 PMCID: PMC4865027 DOI: 10.1186/s12891-016-1060-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/03/2016] [Indexed: 11/10/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked muscle disease that leads to fibre necrosis and progressive paralysis. At present, DMD remains a lethal disease without any effective treatment, requiring a better understanding of the pathophysiological processes and comprehensive assessment of the newly identified therapeutic strategies. MicroRNAs including members of the muscle-specific myomiR family have been identified as being deregulated in muscle of DMD patients and in mdx mice used as a model for DMD. In recent years, the Golden Retriever muscular dystrophy (GRMD) dog has appeared as the crucial animal model for objectively assessing the potential of new innovative approaches. Here, we first aim at establishing the muscle expression pattern of five selected miRNAs in this clinically relevant model to determine if they are similarly affected compared with other DMD contexts. Second, we attempt to show whether these miRNAs could be impacted by the systemic delivery of a promising stem cell candidate (referred to as MuStem cells) to implement our knowledge on its mode of action and/or identify markers associated with cell therapy efficacy. Methods A comparative study of miRNAs expression levels and cellular localization was performed on 9-month-old healthy dogs, as well as on three sub-sets of GRMD dog (without immunosuppression or cell transplantation, with continuous immunosuppressive regimen and with MuStem cell transplantation under immunosuppression), using RT-qPCR and in situ hybridization. Results We find that miR-222 expression is markedly up-regulated in GRMD dog muscle compared to healthy dog, while miR-486 tends to be down-expressed. Intriguingly, the expression of miR-1, miR-133a and miR-206 does not change. In situ hybridization exploration reveals, for the first time, that miR-486 and miR-206 are mainly localized in newly regenerated fibres in GRMD dog muscle. In addition, we show that cyclosporine-based immunosuppression, classically used in allogeneic cell transplantation, exclusively impacts the miR-206 expression. Finally, we demonstrate that intra-arterial administration of MuStem cells results in up-regulation of miR-133a and miR-222 concomitantly with a down-expression of two sarcomeric proteins corresponding to miR-222 targets. Conclusion We point out a differential muscle expression of miR-222 and miR-486 associated with the pathophysiology of the clinically relevant GRMD dog model with a tissue localization focused on regenerated fibres. We also establish a modified expression of miR-133a and miR-222 subsequent to MuStem cell infusion. Electronic supplementary material The online version of this article (doi:10.1186/s12891-016-1060-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Florence Robriquet
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France.,Université de Nantes, F-44322, Nantes, France
| | - Candice Babarit
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Thibaut Larcher
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Laurence Dubreil
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Mireille Ledevin
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Hélicia Goubin
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Karl Rouger
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France.,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France
| | - Laëtitia Guével
- INRA UMR 703 PAnTher "Physiopathologie Animale et bioThérapie du muscle et du système nerveux", Oniris, Atlanpôle - La Chantrerie, Route du Gachet C.S. 40706, F-44307, Nantes Cedex 03, France. .,LUNAM Université, Oniris, École nationale vétérinaire, agro-alimentaire et de l'alimentation Nantes-Atlantique, F-44307, Nantes, France. .,Université de Nantes, F-44322, Nantes, France.
| |
Collapse
|
62
|
Becker S, Florian A, Patrascu A, Rösch S, Waltenberger J, Sechtem U, Schwab M, Schaeffeler E, Yilmaz A. Identification of cardiomyopathy associated circulating miRNA biomarkers in patients with muscular dystrophy using a complementary cardiovascular magnetic resonance and plasma profiling approach. J Cardiovasc Magn Reson 2016; 18:25. [PMID: 27150296 PMCID: PMC4858897 DOI: 10.1186/s12968-016-0244-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/21/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Duchenne and Becker muscular dystrophy (DMD and BMD) are X-chromosomal recessive neuromuscular disorders that are caused by mutations in the dystrophin gene and characterized by cardiac involvement. Circulating microRNAs (miRNAs) have been proposed as diagnostic biomarkers for various cardiovascular diseases. However, circulating miRNAs reflecting the presence and/or disease severity of cardiac involvement in DMD/BMD patients have not been described so far. METHODS Sixty-three male patients with known MD and 26 age-matched healthy male controls were prospectively enrolled. All MD patients and controls underwent comprehensive cardiovascular magnetic resonance (CMR) studies as well as venous blood sampling on the same day. RESULTS An impaired left ventricular (LV) systolic function (defined as LV-EF <55 %) was detected in 29 (46 %) and presence of late gadolinium enhancement (LGE) indicative of myocardial fibrosis in 48 (76 %) MD patients with an exclusively non-ischemic pattern. Whereas no significant differences were observed for the 27 selected circulating miRNAs in MD patients with abnormal CMR findings (comprising structural and/or functional impairments) compared to those with completely normal CMR studies, a significant up-regulation of three miRNAs was observed in LGE-positive MD patients compared to LGE-negative ones: miR-222 (1.8-fold, p = 0.035), miR-26a (2.1-fold, p = 0.03) and miR-378a-5p (2.4-fold, p = 0.026). A signature of these three miRNAs (miR-26a, miR-222 and miR-378a-5p) resulted in an area under the curve (AUC) value of 0.74 for the diagnosis of LGE-positive MD patients. In a multivariable model, three independent predictors for LGE presence were identified comprising not only clinical and laboratory markers (LV-EF: OR 0.47, 95 % CI 0.24-0.89, p = 0.021 and elevated hs-Trop: OR 2559, 95 % CI 2.97-22.04*10(5), p = 0.023) but also the circulating miR-222 (OR 938, 95 % CI 938.46, 3.56-24.73*10(4), p = 0.016). CONCLUSIONS Up-regulation of circulating miRNAs miR-222, miR-26a and miR-378a-5p indicates the presence of myocardial scars in MD patients. Plasma miR-222 appears to be a promising novel biomarker reflecting structural - but not functional - cardiac alterations in MD patients.
Collapse
Affiliation(s)
- Svetlana Becker
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Anca Florian
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | | | - Sabine Rösch
- Division of Cardiology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany
| | - Udo Sechtem
- Division of Cardiology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
- Department of Biochemistry and Pharmacy, University Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Ali Yilmaz
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, building A1, 48149, Münster, Germany.
| |
Collapse
|
63
|
Lidbury J. Potential of circulating microRNAs as biomarkers in veterinary medicine. Vet J 2016; 212:78-9. [PMID: 27256029 DOI: 10.1016/j.tvjl.2016.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Jonathan Lidbury
- Gastrointestinal Laboratory; Texas A&M University; College Station, TX 77843, USA.
| |
Collapse
|
64
|
Hathout Y, Seol H, Han MHJ, Zhang A, Brown KJ, Hoffman EP. Clinical utility of serum biomarkers in Duchenne muscular dystrophy. Clin Proteomics 2016; 13:9. [PMID: 27051355 PMCID: PMC4820909 DOI: 10.1186/s12014-016-9109-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/16/2016] [Indexed: 12/14/2022] Open
Abstract
Assessments of disease progression and response to therapies in Duchenne muscular dystrophy (DMD) patients remain challenging. Current DMD patient assessments include complex physical tests and invasive procedures such as muscle biopsies, which are not suitable for young children. Defining alternative, less invasive and objective outcome measures to assess disease progression and response to therapy will aid drug development and clinical trials in DMD. In this review we highlight advances in development of non-invasive blood circulating biomarkers as a means to assess disease progression and response to therapies in DMD.
Collapse
Affiliation(s)
- Yetrib Hathout
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| | - Haeri Seol
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| | - Meng Hsuan J Han
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| | - Aiping Zhang
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| | - Kristy J Brown
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| | - Eric P Hoffman
- Center for Genetic Medicine, Children's National Healthy System, Washington, DC USA
| |
Collapse
|
65
|
Siracusa J, Koulmann N, Bourdon S, Goriot ME, Banzet S. Circulating miRNAs as Biomarkers of Acute Muscle Damage in Rats. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1313-27. [PMID: 26952641 DOI: 10.1016/j.ajpath.2016.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 01/10/2016] [Accepted: 01/12/2016] [Indexed: 01/15/2023]
Abstract
Skeletal muscle damage is an often-occurring event. Diagnosis using the classic blood marker creatine kinase sometimes yields unsatisfactory results due to great interindividual variability. Therefore, the identification of reliable biomarkers is important. Our aim was to detect and characterize circulating miRNAs in plasma in response to acute notexin-induced muscle damage in rats. Real-time quantitative RT-PCR profiling led to the identification of miRNAs that were highly increased in plasma in response to notexin injection into several muscles, namely miR-1-3p, -133a-3p, -133b-3p, -206-3p, -208b-3p, and -499-5p, as well as miR-378a-3p and miR-434-3p. Peak values of miRNAs appeared 12 hours after injury, and were contained both in the vesicular and nonvesicular fractions of plasma. Receiver operating characteristic curve analysis showed that circulating miRNAs could accurately discriminate between damaged and nondamaged tissues. Furthermore, we tested the robustness of expression profiles in slow- and fast-type fibers. Upon inducing damage in slow- or fast-type muscle, we found that the damaged-muscle phenotype had a very limited impact on the miRNA response. Similarly, the circulating miRNAs selected were not affected by hemolysis or platelets, two pre-analytical factors known to affect plasma miRNA profiles. Taken together, our results show that circulating muscle-specific miRNAs, miR-378a-3p and miR-434-3p, are robust and promising biomarkers of acute muscle damage in rats.
Collapse
Affiliation(s)
- Julien Siracusa
- Armed Forces Biomedical Research Institute, Brétigny-sur-Orge, France
| | - Nathalie Koulmann
- Armed Forces Biomedical Research Institute, Brétigny-sur-Orge, France; Ecole du Val-de-Grâce, Paris, France
| | - Stéphanie Bourdon
- Armed Forces Biomedical Research Institute, Brétigny-sur-Orge, France
| | - Marie-Emmanuelle Goriot
- Armed Forces Biomedical Research Institute/Armed Forces Blood Transfusion Center Jean Julliard, Clamart, France; INSERM U 1197, Clamart, France
| | - Sébastien Banzet
- Armed Forces Biomedical Research Institute/Armed Forces Blood Transfusion Center Jean Julliard, Clamart, France; INSERM U 1197, Clamart, France.
| |
Collapse
|
66
|
Wang Q, Yin BC, Ye BC. A novel polydopamine-based chemiluminescence resonance energy transfer method for microRNA detection coupling duplex-specific nuclease-aided target recycling strategy. Biosens Bioelectron 2016; 80:366-372. [PMID: 26866561 DOI: 10.1016/j.bios.2016.02.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs), functioning as oncogenes or tumor suppressors, play significant regulatory roles in regulating gene expression and become as biomarkers for disease diagnostics and therapeutics. In this work, we have coupled a polydopamine (PDA) nanosphere-assisted chemiluminescence resonance energy transfer (CRET) platform and a duplex-specific nuclease (DSN)-assisted signal amplification strategy to develop a novel method for specific miRNA detection. With the assistance of hemin, luminol, and H2O2, the horseradish peroxidase (HRP)-mimicking G-rich sequence in the sensing probe produces chemiluminescence, which is quickly quenched by the CRET effect between PDA as energy acceptor and excited luminol as energy donor. The target miRNA triggers DSN to partially degrade the sensing probe in the DNA-miRNA heteroduplex to repeatedly release G-quadruplex formed by G-rich sequence from PDA for the production of chemiluminescence. The method allows quantitative detection of target miRNA in the range of 80 pM-50 nM with a detection limit of 49.6 pM. The method also shows excellent specificity to discriminate single-base differences, and can accurately quantify miRNA in biological samples, with good agreement with the result from a commercial miRNA detection kit. The procedure requires no organic dyes or labels, and is a simple and cost-effective method for miRNA detection for early clinical diagnosis.
Collapse
Affiliation(s)
- Qian Wang
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Bin-Cheng Yin
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai 200237, China.
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science & Technology, Shanghai 200237, China.
| |
Collapse
|
67
|
Lv LY, Wang YZ, Zhang Q, Zang HR, Wang XJ. miR-539 induces cell cycle arrest in nasopharyngeal carcinoma by targeting cyclin-dependent kinase 4. Cell Biochem Funct 2015; 33:534-40. [PMID: 26559153 DOI: 10.1002/cbf.3152] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/17/2015] [Accepted: 10/07/2015] [Indexed: 02/02/2023]
Affiliation(s)
- Ling-yan Lv
- Department of Otolaryngology; Affiliated Liaocheng Second People Hospital, Taishan Medical College; Liaocheng China
| | - Yu-zhi Wang
- Department of Otolaryngology; Affiliated Liaocheng Second People Hospital, Taishan Medical College; Liaocheng China
| | - Qian Zhang
- Department of Spinal Surgery; Affiliated Liaocheng Second People Hospital, Taishan Medical College; Liaocheng China
| | - Hong-rui Zang
- Department of Otolaryngology; Beijing Tongren Hospital; Beijing China
| | - Xing-jie Wang
- Department of Otolaryngology; Affiliated Liaocheng Second People Hospital, Taishan Medical College; Liaocheng China
| |
Collapse
|
68
|
Keller A, Leidinger P, Meese E, Haas J, Backes C, Rasche L, Behrens JR, Pfuhl C, Wakonig K, Gieß RM, Jarius S, Meder B, Bellmann-Strobl J, Paul F, Pache FC, Ruprecht K. Next-generation sequencing identifies altered whole blood microRNAs in neuromyelitis optica spectrum disorder which may permit discrimination from multiple sclerosis. J Neuroinflammation 2015; 12:196. [PMID: 26521232 PMCID: PMC4628234 DOI: 10.1186/s12974-015-0418-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 10/21/2015] [Indexed: 01/23/2023] Open
Abstract
Background Neuromyelitis optica spectrum disorder (NMOSD) and multiple sclerosis (MS) have a similar clinical phenotype but represent distinct diseases, requiring different therapies. MicroRNAs (miRNAs) are short non-coding RNAs whose expression profiles can serve as diagnostic biomarkers and which may be involved in the pathophysiology of neuroinflammatory diseases. Here, we analyzed miRNA profiles in serum and whole blood of patients with NMOSD and clinically isolated syndrome (CIS)/relapsing-remitting MS (RRMS) as well as healthy controls by next-generation sequencing (NGS). Methods MiRNA expression profiles were determined by NGS in sera of patients with aquaporin-4 antibody-positive NMOSD (n = 20), CIS/RRMS (n = 20), and healthy controls (n = 20) and in whole blood of patients with NMOSD (n = 11), CIS/RRMS (n = 60), and healthy controls (n = 43). Differentially expressed miRNAs were calculated by analysis of variance and t tests. All significance values were corrected for multiple testing. Selected miRNAs were validated in whole blood of patients with NMOSD (n = 18) and CIS/RRMS (n = 19) by quantitative real-time polymerase chain reaction (qRT-PCR). Results None of 261 miRNAs detected in serum but 178 of 416 miRNAs detected in whole blood showed significantly different expression levels among the three groups. Pairwise comparisons revealed 115 (NMOSD vs. CIS/RRMS), 141 (NMOSD vs. healthy controls), and 44 (CIS/RRMS vs. healthy controls) miRNAs in whole blood with significantly different expression levels. qRT-PCR confirmed different expression levels in whole blood of patients with NMOSD and CIS/RRMS for 9 out of 10 exemplarily chosen miRNAs. In silico enrichment analysis demonstrated an accumulation of altered miRNAs in NMOSD in particular in CD15+ cells (i.e., neutrophils and eosinophils). Conclusions This study identifies a set of miRNAs in whole blood, which may have the potential to discriminate NMOSD from CIS/RRMS and healthy controls. In contrast, miRNA profiles in serum do not appear to be promising diagnostic biomarkers for NMOSD. Enrichment of altered miRNAs in CD15+ neutrophils and eosinophils, which were previously implicated in the pathophysiology of NMOSD, suggests that miRNAs could be involved in the regulation of these cells in NMOSD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0418-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany.
| | | | - Eckart Meese
- Human Genetics, Saarland University, Homburg, Germany.
| | - Jan Haas
- Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.
| | - Christina Backes
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany.
| | - Ludwig Rasche
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Janina R Behrens
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Catherina Pfuhl
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Katharina Wakonig
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - René M Gieß
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | - Benjamin Meder
- Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.
| | - Judith Bellmann-Strobl
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Friedemann Paul
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Florence C Pache
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Klemens Ruprecht
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Clinical and Experimental Multiple Sclerosis Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
69
|
Fiorillo AA, Heier CR, Novak JS, Tully CB, Brown KJ, Uaesoontrachoon K, Vila MC, Ngheim PP, Bello L, Kornegay JN, Angelini C, Partridge TA, Nagaraju K, Hoffman EP. TNF-α-Induced microRNAs Control Dystrophin Expression in Becker Muscular Dystrophy. Cell Rep 2015; 12:1678-90. [PMID: 26321630 DOI: 10.1016/j.celrep.2015.07.066] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/28/2015] [Accepted: 07/29/2015] [Indexed: 12/22/2022] Open
Abstract
The amount and distribution of dystrophin protein in myofibers and muscle is highly variable in Becker muscular dystrophy and in exon-skipping trials for Duchenne muscular dystrophy. Here, we investigate a molecular basis for this variability. In muscle from Becker patients sharing the same exon 45-47 in-frame deletion, dystrophin levels negatively correlate with microRNAs predicted to target dystrophin. Seven microRNAs inhibit dystrophin expression in vitro, and three are validated in vivo (miR-146b/miR-374a/miR-31). microRNAs are expressed in dystrophic myofibers and increase with age and disease severity. In exon-skipping-treated mdx mice, microRNAs are significantly higher in muscles with low dystrophin rescue. TNF-α increases microRNA levels in vitro whereas NFκB inhibition blocks this in vitro and in vivo. Collectively, these data show that microRNAs contribute to variable dystrophin levels in muscular dystrophy. Our findings suggest a model where chronic inflammation in distinct microenvironments induces pathological microRNAs, initiating a self-sustaining feedback loop that exacerbates disease progression.
Collapse
Affiliation(s)
- Alyson A Fiorillo
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Christopher R Heier
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - James S Novak
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Christopher B Tully
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Kristy J Brown
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
| | - Kitipong Uaesoontrachoon
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Maria C Vila
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Peter P Ngheim
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Luca Bello
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Fondazione Ospedale S. Camillo, IRCCS, Lido Venice 30126, Italy
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77845, USA
| | | | - Terence A Partridge
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA.
| |
Collapse
|
70
|
Abstract
Duchenne muscular dystrophy is the most common form of muscular dystrophy. Genetic and biochemical research over the years has characterized the cause, pathophysiology and development of the disease providing several potential therapeutic targets and/or biomarkers. High throughput - omic technologies have provided a comprehensive understanding of the changes occurring in dystrophic muscles. Murine and canine animal models have been a valuable source to profile muscles and body fluids, thus providing candidate biomarkers that can be evaluated in patients. This review will illustrate known circulating biomarkers that could track disease progression and response to therapy in patients affected by Duchenne muscular dystrophy. We present an overview of the transcriptomic, proteomic, metabolomics and lipidomic biomarkers described in literature. We show how studies in muscle tissue have led to the identification of serum and urine biomarkers and we highlight the importance of evaluating biomarkers as possible surrogate endpoints to facilitate regulatory processes for new medicinal products.
Collapse
Affiliation(s)
- Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Human Genetics, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
71
|
Noncoding RNAs, Emerging Regulators of Skeletal Muscle Development and Diseases. BIOMED RESEARCH INTERNATIONAL 2015; 2015:676575. [PMID: 26258142 PMCID: PMC4516831 DOI: 10.1155/2015/676575] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 02/07/2023]
Abstract
A healthy and independent life requires skeletal muscles to maintain optimal function throughout the lifespan, which is in turn dependent on efficient activation of processes that regulate muscle development, homeostasis, and metabolism. Thus, identifying mechanisms that modulate these processes is of crucial priority. Noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have emerged as a class of previously unrecognized transcripts whose importance in a wide range of biological processes and human disease is only starting to be appreciated. In this review, we summarize the roles of recently identified miRNAs and lncRNAs during skeletal muscle development and pathophysiology. We also discuss several molecular mechanisms of these noncoding RNAs. Undoubtedly, further systematic understanding of these noncoding RNAs' functions and mechanisms will not only greatly expand our knowledge of basic skeletal muscle biology, but also significantly facilitate the development of therapies for various muscle diseases, such as muscular dystrophies, cachexia, and sarcopenia.
Collapse
|
72
|
Regulation of smooth muscle contractility by competing endogenous mRNAs in intracranial aneurysms. J Neuropathol Exp Neurol 2015; 74:411-24. [PMID: 25868147 DOI: 10.1097/nen.0000000000000185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alterations in vascular smooth muscle cells (SMCs) contribute to the pathogenesis of intracranial aneurysms (IAs), but the genetic mechanisms underlying these alterations are unclear. We used microarray analysis to compare tissue small noncoding RNA and messenger RNA expression profiles in vessel wall samples from patients with late-stage IAs. We identified myocardin (MYOCD), a key contractility regulator of vascular SMCs, as a critical factor in IA progression. Using a multifaceted computational and experimental approach, we determined that depletion of competitive endogenous RNAs (ARHGEF12, FGF12, and ADCY5) enhanced factors that downregulate MYOCD, which induces the conversion of SMCs from differentiated contractile states into dedifferentiated phenotypes that exhibit enhanced proliferation, synthesis of new extracellular matrix, and organization of mural thrombi. These effects may lead to the repair and maintenance of IAs. This study presents guidelines for the prediction and validation of the IA regulator MYOCD in competitive endogenous RNA networks and facilitates the development of novel therapeutic and diagnostic tools for IAs.
Collapse
|
73
|
Ferlini A, Flanigan KM, Lochmuller H, Muntoni F, 't Hoen PAC, McNally E. 204th ENMC International Workshop on Biomarkers in Duchenne Muscular Dystrophy 24-26 January 2014, Naarden, The Netherlands. Neuromuscul Disord 2015; 25:184-98. [PMID: 25529833 PMCID: PMC4534085 DOI: 10.1016/j.nmd.2014.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/01/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Alessandra Ferlini
- Section of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.
| | - Kevin M Flanigan
- Center for Gene Therapy, Nationwide Children's Hospital, Departments of Pediatrics and Neurology, Ohio State University, Columbus, OH, USA
| | - Hanns Lochmuller
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Francesco Muntoni
- UCL Institute of Child Health & Great Ormond Street Hospital for Children, London, UK
| | - Peter A C 't Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Elizabeth McNally
- Department of Medicine, Department of Human Genetics, The University of Chicago, USA
| |
Collapse
|
74
|
Krist B, Florczyk U, Pietraszek-Gremplewicz K, Józkowicz A, Dulak J. The Role of miR-378a in Metabolism, Angiogenesis, and Muscle Biology. Int J Endocrinol 2015; 2015:281756. [PMID: 26839547 PMCID: PMC4709675 DOI: 10.1155/2015/281756] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
MicroRNA-378a (miR-378a, previously known as miR-378) is one of the small noncoding RNA molecules able to regulate gene expression at posttranscriptional level. Its two mature strands, miR-378a-3p and miR-378a-5p, originate from the first intron of the peroxisome proliferator-activated receptor gamma, coactivator 1 beta (ppargc1b) gene encoding PGC-1β. Embedding in the sequence of this transcriptional regulator of oxidative energy metabolism implies involvement of miR-378a in metabolic pathways, mitochondrial energy homeostasis, and related biological processes such as muscle development, differentiation, and regeneration. On the other hand, modulating the expression of proangiogenic factors such as vascular endothelial growth factor, angiopoietin-1, or interleukin-8, influencing inflammatory reaction, and affecting tumor suppressors, such as SuFu and Fus-1, miR-378a is considered as a part of an angiogenic network in tumors. In the latter, miR-378a can evoke broader actions by enhancing cell survival, reducing apoptosis, and promoting cell migration and invasion. This review describes the current knowledge on miR-378a linking oxidative/lipid metabolism, muscle biology, and blood vessel formation.
Collapse
Affiliation(s)
- Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
- *Jozef Dulak:
| |
Collapse
|