1
|
Kuntawala DH, Vitorino R, Cruz AC, Martins F, Rebelo S. Multisystem Symptoms in Myotonic Dystrophy Type 1: A Management and Therapeutic Perspective. Int J Mol Sci 2025; 26:5350. [PMID: 40508159 PMCID: PMC12155408 DOI: 10.3390/ijms26115350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/20/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a complex, multisystemic neuromuscular disorder with several pathological phenotypes, disease severities and ages of onset. DM1 presents significant challenges in clinical management due to its multisystemic nature, affecting multiple organs and systems beyond skeletal muscle. Tackling this condition requires a comprehensive approach that goes beyond symptom management, particularly considering the complexity of its manifestations and in the delayed diagnosis. In this review we will discuss the multisystem symptoms of DM1 and how this understanding is guiding the development of potential therapies for the improvement of patient outcomes and quality of life. This review aims to explore the available treatments and potential novel disease-modifying therapies targeting DM1 molecular mechanisms to address the broad multisystem symptoms of DM1. Effective strategies to manage symptoms remain crucial, such as physical therapy, medications for myotonia and diligent cardiac care. Metabolic management and hormonal therapies play crucial roles in addressing endocrine and metabolic abnormalities. Nevertheless, promising targeted therapies that include antisense oligonucleotides (ASOs) for RNA degradation, small molecules to disrupt protein-RNA interactions and gene editing offer a prospective approach to the underlying mechanisms of DM1 and improve patient outcomes across the different organ systems.
Collapse
Affiliation(s)
| | | | | | | | - Sandra Rebelo
- Medical Sciences Department, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-183 Aveiro, Portugal; (D.H.K.); (R.V.); (A.C.C.); (F.M.)
| |
Collapse
|
2
|
Lalunio H, Stupka N, Goodman CA, Hayes A. The Potential of Targeting APE1/Ref-1 as a Therapeutic Intervention for Duchenne Muscular Dystrophy. Antioxid Redox Signal 2025; 42:641-654. [PMID: 39729027 DOI: 10.1089/ars.2024.0620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Significance: Inflammation and oxidative stress play crucial roles in the development and progression of skeletal muscle diseases. This review aims to examine the existing evidence regarding the involvement and inhibition of APE1/Ref-1 (apurinic/apyrimidinic endonuclease 1/redox factor 1) in diseases, then extrapolate this evidence to the context of skeletal muscle and discuss the potential beneficial effects of APE1/Ref-1 inhibition in ameliorating myopathy with a particular focus on dystrophic pathology. Critical Issues: Currently, therapeutic interventions targeting pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor erythroid 2-related factor 2 (NRF2), have shown limited efficacy in both clinical and preclinical settings. Thus, there is a need for a more comprehensive treatment approach. Recent Advances: APE1/Ref-1 is a multifunctional protein that was initially identified as being involved in DNA repair. However, newer research has revealed its additional role as a redox-sensitive regulator of transcription factors, including NF-κB and NRF2. Numerous studies have reported increased expression of APE1/Ref-1 in various disorders and have demonstrated the beneficial effects of inhibiting its redox function using the small molecular inhibitor, APX3330. Although these pathways are similarly dysregulated in neuromuscular disorders, the specific role of APE1/Ref-1 in skeletal muscle remains unclear, with only a limited number of studies noting its presence in this tissue. Future Directions: Further studies investigating the role of APE1/Ref-1 in skeletal muscle and identifying whether APE1/Ref-1 is up- or downregulated in dystrophic skeletal muscle would be required to determine whether upregulating or inhibiting the redox function of APE1/Ref-1 will alleviate chronic inflammation and heightened oxidative stress. Antioxid. Redox Signal. 42, 641-654.
Collapse
Affiliation(s)
- Hannah Lalunio
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
| | - Nicole Stupka
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
| | - Craig A Goodman
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
- Institute of Health and Sport (IHeS), Victoria University, Melbourne, Australia
- Centre for Muscle Research, The University of Melbourne, Parkville, Australia
| | - Alan Hayes
- Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St. Albans, Australia
- Institute of Health and Sport (IHeS), Victoria University, Melbourne, Australia
| |
Collapse
|
3
|
Carrascosa-Sàez M, Colom-Rodrigo A, González-Martínez I, Pérez-Gómez R, García-Rey A, Piqueras-Losilla D, Ballestar A, Llamusí B, Cerro-Herreros E, Artero R. Use of HSA LR female mice as a model for the study of myotonic dystrophy type I. Lab Anim (NY) 2025; 54:92-102. [PMID: 40016516 PMCID: PMC11957995 DOI: 10.1038/s41684-025-01506-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/03/2025] [Indexed: 03/01/2025]
Abstract
HSALR mice are the most broadly used animal model for studying myotonic dystrophy type I (DM1). However, so far, HSALR preclinical studies have often excluded female mice or failed to document the biological sex of the animals. This leaves an unwanted knowledge gap concerning the differential development of DM1 in males and females, particularly considering that the disease has a different clinical presentation in men and women. Here we compared typical functional measurements, histological features, molecular phenotypes and biochemical plasma profiles in the muscles of male and female HSALR mice in search of any significant between-sex differences that could justify this exclusion of female mice in HSALR studies and, critically, in candidate therapy assays performed with this model. We found no fundamental differences between HSALR males and females during disease development. Both sexes presented comparable functional and tissue phenotypes, with similar molecular muscle profiles. The only sex differences and significant interactions observed were in plasma biochemical parameters, which are also intrinsically variable in patients with DM1. In addition, we tested the influence of age on these measurements. We therefore suggest including female HSALR mice in regular DM1 studies, and recommend documenting the sex of animals, especially in studies focusing on metabolic alterations. This will allow researchers to detect and report any potential differences between male and female HSALR mice, especially regarding the efficacy of experimental treatments that could be relevant to patients with DM1.
Collapse
Affiliation(s)
- Marc Carrascosa-Sàez
- ARTHEx Biotech, Paterna, Spain
- Institute for Integrative Systems Biology, Consejo Superior de Investigaciones Científicas-Universitat de València, Paterna, Spain
| | - Anna Colom-Rodrigo
- ARTHEx Biotech, Paterna, Spain
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
| | - Irene González-Martínez
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
| | - Raquel Pérez-Gómez
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
| | - Andrea García-Rey
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
- ARTHEx Biotech, Paterna, Spain
| | | | - Ana Ballestar
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
| | | | - Estefanía Cerro-Herreros
- ARTHEx Biotech, Paterna, Spain.
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain.
- Incliva Biomedical Research Institute, Valencia, Spain.
| | - Ruben Artero
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Human Translational Genomics Group, University Institute of Biotechnology and Biomedicine, Universidad de Valencia, Burjassot, Spain
- Incliva Biomedical Research Institute, Valencia, Spain
| |
Collapse
|
4
|
Jennings K, Lindquist D, Poonia A, Schoser B, Schneider-Gold C, Timchenko NA, Timchenko L. The role of CNBP in brain atrophy and its targeting in myotonic dystrophy type 2. Hum Mol Genet 2025; 34:512-522. [PMID: 39807631 DOI: 10.1093/hmg/ddaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Myotonic Dystrophy type 2 (DM2) is a multisystem disease affecting many tissues, including skeletal muscle, heart, and brain. DM2 is caused by unstable expansion of CCTG repeats in an intron 1 of a gene coding for cellular nuclear binding protein (CNBP). The expanded CCTG repeats cause DM2 pathology due to the accumulation of RNA CCUG repeats, which affect RNA processing in patients' cells. We have previously shown that mutant CCUG repeats reduce CNBP protein in DM2 patients. Reducing Cnbp in Cnbp KO mouse model causes late skeletal muscle atrophy. In this study, we examined if the reduction of Cnbp affects the Central Nervous System (CNS). MRI and DTI analyses showed that total brain volume and grey matter are reduced in Cnbp KO mice, while mean, radial and axonal brain diffusivity is increased. The morphological changes in the brains of Cnbp KO mice are accompanied by reduced stereotypic behavior, anxiety and neuromotor defects. These findings suggest that the reduction of CNBP contributes to CNS pathology in DM2. Since CNBP stability is regulated by pAMPK-dependent phosphorylation, we examined protein levels of pAMPK in DM2 cells and found that the active pAMPK is reduced in DM2. Interaction of CNBP with pAMPK and stability of CNBP protein are also decreased in DM2. Our data show that a small molecule AMPK activator A769662 corrects CNBP stability and normalizes CNBP targets in DM2 fibroblasts. Thus, activators of AMPK could potentially be developed as therapeutics to correct CNBP and reduce muscle and brain atrophies in DM2.
Collapse
Affiliation(s)
- Katherine Jennings
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Diana Lindquist
- Imaging Research Center, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
- Departments of Radiology, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
| | - Ankita Poonia
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Benedikt Schoser
- Department of Neurology, Friedrich-Baur-Institute, LMU Clinics, Ziemssenstr, 1 Munich 80336, Germany
| | - Christiane Schneider-Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Gudrinstr. 56, Bochum 44791, Germany
| | - Nikolai A Timchenko
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
- Department of Surgery, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Lubov Timchenko
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
| |
Collapse
|
5
|
Goel K, Saraogi I. Harnessing RNA-Protein Interactions for Therapeutic Interventions. Chem Asian J 2025; 20:e202401117. [PMID: 39714962 DOI: 10.1002/asia.202401117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Interactions between RNAs and proteins play a crucial role in various diseases, including viral infections and cancer. Hence, understanding and inhibiting these interactions are important for the development of novel therapeutics. However, the identification of drugs targeting RNA-protein interactions with high specificity and affinity is challenged by our limited molecular understanding of these interactions. Recent focus on structural and biochemical characterization, coupled with high-throughput screening technologies and computational modeling, have accelerated the identification of new RBPs and optimization of potential inhibitors. This review discusses key examples of inhibitors developed over the past decade that effectively disrupt pathogenic RNA-protein interactions. We focus on small molecule and peptide-based inhibitors that have shown promise in disrupting crucial RNA-protein interactions in eukaryotes, prokaryotes, and viruses. We also present the challenges and future directions in this field, emphasizing the need to achieve improved specificity and reduce the off-target effects of the inhibitors. This review aims to contribute to ongoing efforts towards the development of novel therapeutic agents targeting RNA-protein interactions by providing an in-depth analysis of significant developments and emerging trends in this rapidly growing field.
Collapse
Affiliation(s)
- Khushboo Goel
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Ishu Saraogi
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal Bhauri, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
6
|
Hicks SM, Frias JA, Mishra SK, Scotti M, Muscato DR, Valero MC, Adams LM, Cleary JD, Nakamori M, Wang E, Berglund JA. Alternative splicing dysregulation across tissue and therapeutic approaches in a mouse model of myotonic dystrophy type 1. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102338. [PMID: 39391766 PMCID: PMC11465180 DOI: 10.1016/j.omtn.2024.102338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024]
Abstract
Myotonic dystrophy type 1 (DM1), the leading cause of adult-onset muscular dystrophy, is caused by a CTG repeat expansion. Expression of the repeat causes widespread alternative splicing (AS) defects and downstream pathogenesis, including significant skeletal muscle impacts. The HSA LR mouse model plays a significant role in therapeutic development. This mouse model features a transgene composed of approximately 220 interrupted CTG repeats, which results in skeletal muscle pathology that mirrors DM1. To better understand this model and the growing number of therapeutic approaches developed with it, we performed a meta-analysis of publicly available RNA sequencing data for AS changes across three widely examined skeletal muscles: quadriceps, gastrocnemius, and tibialis anterior. Our analysis demonstrated that transgene expression correlated with the extent of splicing dysregulation across these muscles from gastrocnemius (highest), quadriceps (medium), to tibialis anterior (lowest). We identified 95 splicing events consistently dysregulated across all examined datasets. Comparison of splicing rescue across seven therapeutic approaches showed a range of rescue across the 95 splicing events from the three muscle groups. This analysis contributes to our understanding of the HSA LR model and the growing number of therapeutic approaches currently in preclinical development for DM1.
Collapse
Affiliation(s)
- Sawyer M. Hicks
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Jesus A. Frias
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Subodh K. Mishra
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Marina Scotti
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - Derek R. Muscato
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - M. Carmen Valero
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - Leanne M. Adams
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - John D. Cleary
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eric Wang
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - J. Andrew Berglund
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| |
Collapse
|
7
|
Marcella BM, Hockey BL, Braun JL, Whitley KC, Geromella MS, Baranowski RW, Watson CJF, Silvera S, Hamstra SI, Wasilewicz LJ, Crozier RWE, Marais AAT, Kim KH, Lee G, Vandenboom R, Roy BD, MacNeil AJ, MacPherson REK, Fajardo VA. GSK3 inhibition improves skeletal muscle function and whole-body metabolism in male mouse models of Duchenne muscular dystrophy. Nat Commun 2024; 15:10210. [PMID: 39587049 PMCID: PMC11589163 DOI: 10.1038/s41467-024-53886-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/25/2024] [Indexed: 11/27/2024] Open
Abstract
Inhibiting glycogen synthase kinase 3 (GSK3) improves muscle function, metabolism, and bone health in many diseases and conditions; however, whether GSK3 should be targeted for Duchenne muscular dystrophy (DMD), a severe muscle wasting disorder with no cure, remains unknown. Here, we show the effects of GSK3 inhibition in male DBA/2J (D2) and C57BL/10 (C57) mdx mice. Treating D2 mdx mice with GSK3 inhibitors alone or in combination with aerobic exercise improves muscle strength, endurance, and morphology, attenuates the hypermetabolic phenotype, and enhances insulin sensitivity. GSK3 inhibition in C57 mdx mice also improves muscle fatigue resistance and increases cage ambulation. Moreover, muscle-specific GSK3 knockdown in mdx mice augments muscle force production and endurance. In both mdx strains, GSK3 inhibition increases bone mineral content and density. Overall, these improvements to muscle, metabolic, and bone health with GSK3 inhibition in mdx mice may have clinical implications for patients with DMD, where the current standard of care, glucocorticoids, delay the loss of ambulation but increase the risk for insulin resistance and osteoporosis. Along with our observation of lowered β-catenin content in DMD myoblasts, a known cellular target for GSK3, this study provides ample evidence in support of inhibiting GSK3 for this disease.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Male
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Mice, Inbred mdx
- Mice, Inbred C57BL
- Mice
- Disease Models, Animal
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3/antagonists & inhibitors
- Muscle Strength/drug effects
- Mice, Inbred DBA
- Physical Conditioning, Animal
- Bone Density/drug effects
- Insulin Resistance
Collapse
Affiliation(s)
- Bianca M Marcella
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Briana L Hockey
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
- Centre for Neurosciences, Brock University, St. Catharines, ON, Canada
| | - Kennedy C Whitley
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Mia S Geromella
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Colton J F Watson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Sebastian Silvera
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Luc J Wasilewicz
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Robert W E Crozier
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Amélie A T Marais
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Kun Ho Kim
- Institute for Cell Engineering Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gabsang Lee
- Institute for Cell Engineering Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rene Vandenboom
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Brian D Roy
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Adam J MacNeil
- Centre for Neurosciences, Brock University, St. Catharines, ON, Canada
| | - Rebecca E K MacPherson
- Centre for Neurosciences, Brock University, St. Catharines, ON, Canada
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada.
- Centre for Neurosciences, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
8
|
Tan Y, Zhao Z, Han Q, Xu P, Shen X, Jiang Y, Xu Q, Wu X. Identification of an RNA-binding perturbing characteristic for thiopurine drugs and their derivatives to disrupt CELF1-RNA interaction. Nucleic Acids Res 2024; 52:10810-10822. [PMID: 39268573 PMCID: PMC11472155 DOI: 10.1093/nar/gkae788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
RNA-binding proteins (RBPs) are attractive targets in human pathologies. Despite a number of efforts to target RBPs with small molecules, it is still difficult to develop RBP inhibitors, asking for a deeper understanding of how to chemically perturb RNA-binding activity. In this study, we found that the thiopurine drugs (6-mercaptopurine and 6-thioguanine) effectively disrupt CELF1-RNA interaction. The disrupting activity relies on the formation of disulfide bonds between the thiopurine drugs and CELF1. Mutating the cysteine residue proximal to the RNA recognition motifs (RRMs), or adding reducing agents, abolishes the disrupting activity. Furthermore, the 1,2,4-triazole-3-thione, a thiopurine analogue, was identified with 20-fold higher disrupting activity. Based on this analogue, we found that compound 9 disrupts CELF1-RNA interaction in living cells and ameliorates CELF1-mediated myogenesis deficiency. In summary, we identified a thiol-mediated binding mechanism for thiopurine drugs and their derivatives to perturb protein-RNA interaction, which provides novel insight for developing RBP inhibitors. Additionally, this work may benefit the pharmacological and toxicity research of thiopurine drugs.
Collapse
Affiliation(s)
- Yang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhibo Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Qingfang Han
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Peipei Xu
- Department of Hematology, Drum Tower Hospital Affiliated to Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaopeng Shen
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Yajun Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Xingxin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Drum Tower Hospital Affiliated to Medical School, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
9
|
DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, Posner BA, Hulleman JD. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. JCI Insight 2024; 9:e178050. [PMID: 39114980 PMCID: PMC11383595 DOI: 10.1172/jci.insight.178050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/20/2024] [Indexed: 08/22/2024] Open
Abstract
Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) is an age-related macular degeneration-like (AMD-like) retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production in retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus that enabled simple, sensitive, and high-throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix, reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium-derived factor). In vivo, treatment of 8-month-old R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is an important demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of retinal degenerative diseases, including potentially AMD itself.
Collapse
Affiliation(s)
- Sophia M. DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Krista N. Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Melissa K. McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
10
|
Moreno N, Sabater-Arcis M, Sevilla T, Alonso MP, Ohana J, Bargiela A, Artero R. Therapeutic potential of oleic acid supplementation in myotonic dystrophy muscle cell models. Biol Res 2024; 57:29. [PMID: 38760841 PMCID: PMC11100173 DOI: 10.1186/s40659-024-00496-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/05/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND We recently reported that upregulation of Musashi 2 (MSI2) protein in the rare neuromuscular disease myotonic dystrophy type 1 contributes to the hyperactivation of the muscle catabolic processes autophagy and UPS through a reduction in miR-7 levels. Because oleic acid (OA) is a known allosteric regulator of MSI2 activity in the biogenesis of miR-7, here we sought to evaluate endogenous levels of this fatty acid and its therapeutic potential in rescuing cell differentiation phenotypes in vitro. In this work, four muscle cell lines derived from DM1 patients were treated with OA for 24 h, and autophagy and muscle differentiation parameters were analyzed. RESULTS We demonstrate a reduction of OA levels in different cell models of the disease. OA supplementation rescued disease-related phenotypes such as fusion index, myotube diameter, and repressed autophagy. This involved inhibiting MSI2 regulation of direct molecular target miR-7 since OA isoschizomer, elaidic acid (EA) could not cause the same rescues. Reduction of OA levels seems to stem from impaired biogenesis since levels of the enzyme stearoyl-CoA desaturase 1 (SCD1), responsible for converting stearic acid to oleic acid, are decreased in DM1 and correlate with OA amounts. CONCLUSIONS For the first time in DM1, we describe a fatty acid metabolism impairment that originated, at least in part, from a decrease in SCD1. Because OA allosterically inhibits MSI2 binding to molecular targets, reduced OA levels synergize with the overexpression of MSI2 and contribute to the MSI2 > miR-7 > autophagy axis that we proposed to explain the muscle atrophy phenotype.
Collapse
Affiliation(s)
- Nerea Moreno
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Maria Sabater-Arcis
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Teresa Sevilla
- CIBERER, IISCIII, Madrid, Spain
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital, La Fe (IIS La Fe), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Manuel Perez Alonso
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| | - Jessica Ohana
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, Institut de Myologie, Paris, F-75013, France
| | - Ariadna Bargiela
- CIBERER, IISCIII, Madrid, Spain.
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital, La Fe (IIS La Fe), Valencia, Spain.
| | - Ruben Artero
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- CIBERER, IISCIII, Madrid, Spain
| |
Collapse
|
11
|
Zambon AA, Falzone YM, Bolino A, Previtali SC. Molecular mechanisms and therapeutic strategies for neuromuscular diseases. Cell Mol Life Sci 2024; 81:198. [PMID: 38678519 PMCID: PMC11056344 DOI: 10.1007/s00018-024-05229-9] [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: 01/02/2024] [Revised: 03/14/2024] [Accepted: 04/07/2024] [Indexed: 05/01/2024]
Abstract
Neuromuscular diseases encompass a heterogeneous array of disorders characterized by varying onset ages, clinical presentations, severity, and progression. While these conditions can stem from acquired or inherited causes, this review specifically focuses on disorders arising from genetic abnormalities, excluding metabolic conditions. The pathogenic defect may primarily affect the anterior horn cells, the axonal or myelin component of peripheral nerves, the neuromuscular junction, or skeletal and/or cardiac muscles. While inherited neuromuscular disorders have been historically deemed not treatable, the advent of gene-based and molecular therapies is reshaping the treatment landscape for this group of condition. With the caveat that many products still fail to translate the positive results obtained in pre-clinical models to humans, both the technological development (e.g., implementation of tissue-specific vectors) as well as advances on the knowledge of pathogenetic mechanisms form a collective foundation for potentially curative approaches to these debilitating conditions. This review delineates the current panorama of therapies targeting the most prevalent forms of inherited neuromuscular diseases, emphasizing approved treatments and those already undergoing human testing, offering insights into the state-of-the-art interventions.
Collapse
Affiliation(s)
- Alberto Andrea Zambon
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Institute for Experimental Neurology, Inspe, Milan, Italy
- Neurology Department, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Yuri Matteo Falzone
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Institute for Experimental Neurology, Inspe, Milan, Italy
- Neurology Department, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Bolino
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Institute for Experimental Neurology, Inspe, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Carlo Previtali
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Institute for Experimental Neurology, Inspe, Milan, Italy.
- Neurology Department, San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
12
|
Yadava RS, Mandal M, Mahadevan MS. Studying the Effect of MBNL1 and MBNL2 Loss in Skeletal Muscle Regeneration. Int J Mol Sci 2024; 25:2687. [PMID: 38473933 PMCID: PMC10931579 DOI: 10.3390/ijms25052687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Loss of function of members of the muscleblind-like (MBNL) family of RNA binding proteins has been shown to play a key role in the spliceopathy of RNA toxicity in myotonic dystrophy type 1 (DM1), the most common muscular dystrophy affecting adults and children. MBNL1 and MBNL2 are the most abundantly expressed members in skeletal muscle. A key aspect of DM1 is poor muscle regeneration and repair, leading to dystrophy. We used a BaCl2-induced damage model of muscle injury to study regeneration and effects on skeletal muscle satellite cells (MuSCs) in Mbnl1∆E3/∆E3 and Mbnl2∆E2/∆E2 knockout mice. Similar experiments have previously shown deleterious effects on these parameters in mouse models of RNA toxicity. Muscle regeneration in Mbnl1 and Mbnl2 knockout mice progressed normally with no obvious deleterious effects on MuSC numbers or increased expression of markers of fibrosis. Skeletal muscles in Mbnl1∆E3/∆E3/ Mbnl2∆E2/+ mice showed increased histopathology but no deleterious reductions in MuSC numbers and only a slight increase in collagen deposition. These results suggest that factors beyond the loss of MBNL1/MBNL2 and the associated spliceopathy are likely to play a key role in the defects in skeletal muscle regeneration and deleterious effects on MuSCs that are seen in mouse models of RNA toxicity due to expanded CUG repeats.
Collapse
Affiliation(s)
| | | | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA; (R.S.Y.)
| |
Collapse
|
13
|
Nakamori M. Expanded‐repeat‐RNA‐mediated disease mechanisms in myotonic dystrophy. NEUROLOGY AND CLINICAL NEUROSCIENCE 2024; 12:16-23. [DOI: 10.1111/ncn3.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/05/2022] [Indexed: 01/04/2025]
Abstract
AbstractMyotonic dystrophy (DM) is the most common muscular dystrophy in adults, affecting skeletal muscle as well as cardiac and smooth muscle. Furthermore, involvement of the central nervous system, endocrine organs, and eyes is often seen, with debilitating consequences. The condition is an autosomal‐dominant inherited genetic disease caused by abnormal genomic expansion of tandem repeats. Myotonic dystrophy type 1 (DM1) results from expansion of a CTG repeat in the 3′‐untranslated region of the gene encoding dystrophia myotonica‐protein kinase (DMPK), whereas myotonic dystrophy type 2 (DM2) is caused by expansion of a CCTG repeat in the first intron of the gene encoding CCHC‐type zinc finger nucleic acid‐binding protein (CNBP). Both types of DM exhibit abnormal mRNA transcribed from the mutated gene containing expanded repeats, which exert toxic gain‐of‐function effects on various proteins involved in cellular processes such as alternative splicing, signaling pathways, and cellular senescence. The present review discusses the expanded‐repeat‐RNA‐mediated molecular pathomechanisms in DM.
Collapse
Affiliation(s)
- Masayuki Nakamori
- Department of Neurology Osaka University Graduate School of Medicine Osaka Japan
| |
Collapse
|
14
|
DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, Posner BA, Hulleman JD. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571757. [PMID: 38168310 PMCID: PMC10760106 DOI: 10.1101/2023.12.14.571757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Malattia Leventinese/Doyne Honeycomb Retinal Dystrophy (ML/DHRD) is an age-related macular degeneration (AMD)-like retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production from retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus which enabled simple, sensitive, and high throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix (ECM), reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium derived factor). In vivo, treatment of 8 mo R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is the first demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of neurodegenerative diseases, including, potentially AMD itself.
Collapse
Affiliation(s)
- Sophia M. DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Antonio J. Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Gracen E. Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| | - Krista N. Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390, United States
| | - Melissa K. McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, United States
| | - Bruce A. Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, United States
| | - John D. Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6 St. SE, Minneapolis, Minnesota, 55455, United States
| |
Collapse
|
15
|
Lutz M, Levanti M, Karns R, Gourdon G, Lindquist D, Timchenko NA, Timchenko L. Therapeutic Targeting of the GSK3β-CUGBP1 Pathway in Myotonic Dystrophy. Int J Mol Sci 2023; 24:10650. [PMID: 37445828 PMCID: PMC10342152 DOI: 10.3390/ijms241310650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Myotonic Dystrophy type 1 (DM1) is a neuromuscular disease associated with toxic RNA containing expanded CUG repeats. The developing therapeutic approaches to DM1 target mutant RNA or correct early toxic events downstream of the mutant RNA. We have previously described the benefits of the correction of the GSK3β-CUGBP1 pathway in DM1 mice (HSALR model) expressing 250 CUG repeats using the GSK3 inhibitor tideglusib (TG). Here, we show that TG treatments corrected the expression of ~17% of genes misregulated in DM1 mice, including genes involved in cell transport, development and differentiation. The expression of chloride channel 1 (Clcn1), the key trigger of myotonia in DM1, was also corrected by TG. We found that correction of the GSK3β-CUGBP1 pathway in mice expressing long CUG repeats (DMSXL model) is beneficial not only at the prenatal and postnatal stages, but also during adulthood. Using a mouse model with dysregulated CUGBP1, which mimics alterations in DM1, we showed that the dysregulated CUGBP1 contributes to the toxicity of expanded CUG repeats by changing gene expression and causing CNS abnormalities. These data show the critical role of the GSK3β-CUGBP1 pathway in DM1 muscle and in CNS pathologies, suggesting the benefits of GSK3 inhibitors in patients with different forms of DM1.
Collapse
Affiliation(s)
- Maggie Lutz
- Division of Neurology, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA; (M.L.); (M.L.)
| | - Miranda Levanti
- Division of Neurology, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA; (M.L.); (M.L.)
| | - Rebekah Karns
- Departments of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA;
| | - Genevieve Gourdon
- Sorbonne Université, Inserm, institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France;
| | - Diana Lindquist
- Imagine Research Center, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA;
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45221, USA;
| | - Nikolai A. Timchenko
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45221, USA;
- Department of Surgery, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA
| | - Lubov Timchenko
- Division of Neurology, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA; (M.L.); (M.L.)
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45221, USA;
| |
Collapse
|
16
|
Pascual-Gilabert M, Artero R, López-Castel A. The myotonic dystrophy type 1 drug development pipeline: 2022 edition. Drug Discov Today 2023; 28:103489. [PMID: 36634841 DOI: 10.1016/j.drudis.2023.103489] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
The beginning of the 20th decade has witnessed an increase in drug development programs for myotonic dystrophy type 1 (DM1). We have collected nearly 20 candidate drugs with accomplished preclinical and clinical phases, updating our previous drug development pipeline review with new entries and relevant milestones for pre-existing candidates. Three interventional first-in-human clinical trials got underway with distinct drug classes, namely AOC 1001 and DYNE-101 nucleic acid-based therapies, and the small molecule pitolisant, which joins the race toward market authorization with other repurposed drugs, including tideglusib, metformin, or mexiletine, already in clinical evaluation. Furthermore, newly disclosed promising preclinical data for several additional nucleic-acid therapeutic candidates and a CRISPR-based approach, as well as the advent into the pipeline of novel therapeutic programs, increase the plausibility of success in the demanding task of providing valid treatments to patients with DM1.
Collapse
Affiliation(s)
| | - Ruben Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain; Translational Genomics Group, Incliva Biomedical Research Institute, Valencia, Spain.
| | - Arturo López-Castel
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain; Translational Genomics Group, Incliva Biomedical Research Institute, Valencia, Spain.
| |
Collapse
|
17
|
Administration of a single dose of lithium ameliorates rhabdomyolysis-associated acute kidney injury in rats. PLoS One 2023; 18:e0281679. [PMID: 36795689 PMCID: PMC9934413 DOI: 10.1371/journal.pone.0281679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Rhabdomyolysis is characterized by muscle damage and leads to acute kidney injury (AKI). Clinical and experimental studies suggest that glycogen synthase kinase 3β (GSK3β) inhibition protects against AKI basically through its critical role in tubular epithelial cell apoptosis, inflammation and fibrosis. Treatment with a single dose of lithium, an inhibitor of GSK3β, accelerated recovery of renal function in cisplatin and ischemic/reperfusion-induced AKI models. We aimed to evaluate the efficacy of a single dose of lithium in the treatment of rhabdomyolysis-induced AKI. Male Wistar rats were allocated to four groups: Sham, received saline 0.9% intraperitoneally (IP); lithium (Li), received a single IP injection of lithium chloride (LiCl) 80 mg/kg body weight (BW); glycerol (Gly), received a single dose of glycerol 50% 5 mL/kg BW intramuscular (IM); glycerol plus lithium (Gly+Li), received a single dose of glycerol 50% IM plus LiCl IP injected 2 hours after glycerol administration. After 24 hours, we performed inulin clearance experiments and collected blood / kidney / muscle samples. Gly rats exhibited renal function impairment accompanied by kidney injury, inflammation and alterations in signaling pathways for apoptosis and redox state balance. Gly+Li rats showed a remarkable improvement in renal function as well as kidney injury score, diminished CPK levels and an overstated decrease of renal and muscle GSK3β protein expression. Furthermore, administration of lithium lowered the amount of macrophage infiltrate, reduced NFκB and caspase renal protein expression and increased the antioxidant component MnSOD. Lithium treatment attenuated renal dysfunction in rhabdomyolysis-associated AKI by improving inulin clearance and reducing CPK levels, inflammation, apoptosis and oxidative stress. These therapeutic effects were due to the inhibition of GSK3β and possibly associated with a decrease in muscle injury.
Collapse
|
18
|
Costa A, Cruz AC, Martins F, Rebelo S. Protein Phosphorylation Alterations in Myotonic Dystrophy Type 1: A Systematic Review. Int J Mol Sci 2023; 24:ijms24043091. [PMID: 36834509 PMCID: PMC9965115 DOI: 10.3390/ijms24043091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Among the most common muscular dystrophies in adults is Myotonic Dystrophy type 1 (DM1), an autosomal dominant disorder characterized by myotonia, muscle wasting and weakness, and multisystemic dysfunctions. This disorder is caused by an abnormal expansion of the CTG triplet at the DMPK gene that, when transcribed to expanded mRNA, can lead to RNA toxic gain of function, alternative splicing impairments, and dysfunction of different signaling pathways, many regulated by protein phosphorylation. In order to deeply characterize the protein phosphorylation alterations in DM1, a systematic review was conducted through PubMed and Web of Science databases. From a total of 962 articles screened, 41 were included for qualitative analysis, where we retrieved information about total and phosphorylated levels of protein kinases, protein phosphatases, and phosphoproteins in DM1 human samples and animal and cell models. Twenty-nine kinases, 3 phosphatases, and 17 phosphoproteins were reported altered in DM1. Signaling pathways that regulate cell functions such as glucose metabolism, cell cycle, myogenesis, and apoptosis were impaired, as seen by significant alterations to pathways such as AKT/mTOR, MEK/ERK, PKC/CUGBP1, AMPK, and others in DM1 samples. This explains the complexity of DM1 and its different manifestations and symptoms, such as increased insulin resistance and cancer risk. Further studies can be done to complement and explore in detail specific pathways and how their regulation is altered in DM1, to find what key phosphorylation alterations are responsible for these manifestations, and ultimately to find therapeutic targets for future treatments.
Collapse
|
19
|
Misquitta NS, Ravel-Chapuis A, Jasmin BJ. Combinatorial treatment with exercise and AICAR potentiates the rescue of myotonic dystrophy type 1 mouse muscles in a sex-specific manner. Hum Mol Genet 2023; 32:551-566. [PMID: 36048859 DOI: 10.1093/hmg/ddac222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023] Open
Abstract
Targeting AMP-activated protein kinase (AMPK) is emerging as a promising strategy for treating myotonic dystrophy type 1 (DM1), the most prevalent form of adult-onset muscular dystrophy. We previously demonstrated that 5-aminomidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) and exercise, two potent AMPK activators, improve disease features in DM1 mouse skeletal muscles. Here, we employed a combinatorial approach with these AMPK activators and examined their joint impact on disease severity in male and female DM1 mice. Our data reveal that swimming exercise additively enhances the effect of AICAR in mitigating the nuclear accumulation of toxic CUGexp RNA foci. In addition, our findings show a trend towards an enhanced reversal of MBNL1 sequestration and correction in pathogenic alternative splicing events. Our results further demonstrate that the combinatorial impact of exercise and AICAR promotes muscle fiber hypertrophy in DM1 skeletal muscle. Importantly, these improvements occur in a sex-specific manner with greater benefits observed in female DM1 mice. Our findings demonstrate that combining AMPK-activating interventions may prove optimal for rescuing the DM1 muscle phenotype and uncover important sex differences in the response to AMPK-based therapeutic strategies in DM1 mice.
Collapse
Affiliation(s)
- Naomi S Misquitta
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,The Eric J. Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,The Eric J. Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,The Eric J. Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
20
|
Quantitative magnetic resonance imaging assessment of muscle composition in myotonic dystrophy mice. Sci Rep 2023; 13:503. [PMID: 36627397 PMCID: PMC9831979 DOI: 10.1038/s41598-023-27661-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a severe autosomal dominant neuromuscular disease in which the musculoskeletal system contributes substantially to overall mortality and morbidity. DM1 stems from a noncoding CTG trinucleotide repeat expansion in the DMPK gene. The human skeletal actin long repeat (HSALR) mouse model reproduces several aspects of the disease, but the muscle-wasting phenotype of this model has never been characterized in vivo. Herein, we used quantitative MRI to measure the fat and muscle volumes in the leg compartment (LC) of mice. These acquired data were processed to extract relevant parameters such as fat fraction and fat infiltration (fat LC/LC) in HSALR and control (FBV) muscles. These results showed increased fat volume (fat LC) and fat infiltration within the muscle tissue of the leg compartment (muscle LC), in agreement with necropsies, in which fatty clumps were observed, and consistent with previous findings in DM1 patients. Model mice did not reproduce the characteristic impaired fat fraction, widespread fat replacement through the muscles, or reduced muscle volume reported in patients. Taken together, the observed abnormal replacement of skeletal muscle by fat in the HSALR mice indicates that these mice partially reproduced the muscle phenotype observed in humans.
Collapse
|
21
|
Development of Therapeutic Approaches for Myotonic Dystrophies Type 1 and Type 2. Int J Mol Sci 2022; 23:ijms231810491. [PMID: 36142405 PMCID: PMC9499601 DOI: 10.3390/ijms231810491] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Myotonic Dystrophies type 1 (DM1) and type 2 (DM2) are complex multisystem diseases without disease-based therapies. These disorders are caused by the expansions of unstable CTG (DM1) and CCTG (DM2) repeats outside of the coding regions of the disease genes: DMPK in DM1 and CNBP in DM2. Multiple clinical and molecular studies provided a consensus for DM1 pathogenesis, showing that the molecular pathophysiology of DM1 is associated with the toxicity of RNA CUG repeats, which cause multiple disturbances in RNA metabolism in patients' cells. As a result, splicing, translation, RNA stability and transcription of multiple genes are misregulated in DM1 cells. While mutant CCUG repeats are the main cause of DM2, additional factors might play a role in DM2 pathogenesis. This review describes current progress in the translation of mechanistic knowledge in DM1 and DM2 to clinical trials, with a focus on the development of disease-specific therapies for patients with adult forms of DM1 and congenital DM1 (CDM1).
Collapse
|
22
|
Ait Benichou S, Jauvin D, De Serres-Bérard T, Bennett F, Rigo F, Gourdon G, Boutjdir M, Chahine M, Puymirat J. Enhanced Delivery of Ligand-Conjugated Antisense Oligonucleotides (C16-HA-ASO) Targeting Dystrophia Myotonica Protein Kinase Transcripts for the Treatment of Myotonic Dystrophy Type 1. Hum Gene Ther 2022; 33:810-820. [PMID: 35794764 DOI: 10.1089/hum.2022.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a neuromuscular disorder that affects many organs. It is caused by the expansion of a cytosine-thymine-guanine triplet repeat in the 3' untranslated region of the human dystrophia myotonica protein kinase (hDMPK) gene, which results in a toxic gain of function of mutant hDMPK RNA transcripts. Antisense oligonucleotides (ASOs) have emerged in recent years as a potential gene therapy to treat DM1. However, the clinical efficacy of the systemic administration of ASOs is limited by a combination of insufficient potency and poor tissue distribution. In the present study, we assessed the potential of a new ligand-conjugated ASO (IONIS-877864; C16-HA-ASO) to target mutant hDMPK mRNA transcripts in the DMSXL mouse model of DM1 carrying over 1000 CTG pathogenic repeats. DMSXL mice were treated subcutaneously for 9 weeks with either IONIS-877864 (12.5 or 25 mg/kg) or IONIS-486178 (12.5 or 25 mg/kg), an unconjugated ASO with the same sequence. At 25 mg/kg, IONIS-877864 significantly enhanced ASO delivery into the striated muscles of DMSXL mice following systemic administration compared with the unconjugated control. IONIS-877864 was also more efficacious than IONIS-486178, reducing mutant hDMPK transcripts by up to 92% in the skeletal muscles and 78% in the hearts of DMSXL mice. The decrease in mutant hDMPK transcripts in skeletal muscles caused by IONIS-877864 was associated with a significant improvement in muscle strength. IONIS-877864 was nontoxic in the DMSXL mouse model. The present study showed that the C16-HA-conjugated ASO is a powerful tool for the development of gene therapy for DM1.
Collapse
Affiliation(s)
- Siham Ait Benichou
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada
| | - Dominic Jauvin
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, Canada
| | - Thiéry De Serres-Bérard
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, Canada
| | - Frank Bennett
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Genevieve Gourdon
- Centre de Recherche en Myologie, Association Institut de Myologie, Sorbonne Université, Inserm, Paris, France
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, New York, USA
- Department of Medicine, Cell Biology, and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Mohamed Chahine
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, Canada
- Department of Medicine, Faculté de Médecine, Université Laval, Quebec City, Canada
| | - Jack Puymirat
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Medicine, Faculté de Médecine, Université Laval, Quebec City, Canada
| |
Collapse
|
23
|
Ravel-Chapuis A, Duchesne E, Jasmin BJ. Pharmacological and exercise-induced activation of AMPK as emerging therapies for myotonic dystrophy type 1 patients. J Physiol 2022; 600:3249-3264. [PMID: 35695045 DOI: 10.1113/jp282725] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/07/2022] [Indexed: 11/08/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disorder with variable clinical features. Currently, there is no cure or effective treatment for DM1. The disease is caused by an expansion of CUG repeats in the 3' UTR of DMPK mRNAs. Mutant DMPK mRNAs accumulate in nuclei as RNA foci and trigger an imbalance in the level and localization of RNA-binding proteins causing the characteristic missplicing events that account for the varied DM1 symptoms, a disease mechanism referred to as RNA toxicity. In recent years, multiple signalling pathways have been identified as being aberrantly regulated in skeletal muscle in response to the CUG expansion, including AMPK, a sensor of energy status, as well as a master regulator of cellular energy homeostasis. Converging lines of evidence highlight the benefits of activating AMPK signalling pharmacologically on RNA toxicity, as well as on muscle histology and function, in preclinical DM1 models. Importantly, a clinical trial with metformin, an activator of AMPK, resulted in functional benefits in DM1 patients. In addition, exercise, a known AMPK activator, has shown promising effects on RNA toxicity and muscle function in DM1 mice. Finally, clinical trials involving moderate-intensity exercise also induced functional benefits for DM1 patients. Taken together, these studies clearly demonstrate the molecular, histological and functional benefits of AMPK activation and exercise-based interventions on the DM1 phenotype. Despite these advances, several key questions remain; in particular, the extent of the true implication of AMPK in the observed beneficial improvements, as well as how, mechanistically, activation of AMPK signalling improves the DM1 pathophysiology.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Elise Duchesne
- Département des sciences de la santé, Université du Québec à Chicoutimi, Chicoutimi, QC, Canada.,Groupe de Recherche Interdisciplinaire sur les Maladies Neuromusculaires (GRIMN), Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Hôpital de Jonquière, QC, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
24
|
Molecular Therapies for Myotonic Dystrophy Type 1: From Small Drugs to Gene Editing. Int J Mol Sci 2022; 23:ijms23094622. [PMID: 35563013 PMCID: PMC9101876 DOI: 10.3390/ijms23094622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy affecting many different body tissues, predominantly skeletal and cardiac muscles and the central nervous system. The expansion of CTG repeats in the DM1 protein-kinase (DMPK) gene is the genetic cause of the disease. The pathogenetic mechanisms are mainly mediated by the production of a toxic expanded CUG transcript from the DMPK gene. With the availability of new knowledge, disease models, and technical tools, much progress has been made in the discovery of altered pathways and in the potential of therapeutic intervention, making the path to the clinic a closer reality. In this review, we describe and discuss the molecular therapeutic strategies for DM1, which are designed to directly target the CTG genomic tract, the expanded CUG transcript or downstream signaling molecules.
Collapse
|
25
|
Mikhail AI, Nagy PL, Manta K, Rouse N, Manta A, Ng SY, Nagy MF, Smith P, Lu JQ, Nederveen JP, Ljubicic V, Tarnopolsky MA. Aerobic exercise elicits clinical adaptations in myotonic dystrophy type 1 patients independent of pathophysiological changes. J Clin Invest 2022; 132:156125. [PMID: 35316212 PMCID: PMC9106360 DOI: 10.1172/jci156125] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is a complex life-limiting neuromuscular disorder characterized by severe skeletal muscle atrophy, weakness, and cardio-respiratory defects. Exercised DM1 mice exhibit numerous physiological benefits that are underpinned by reduced CUG foci and improved alternative splicing. However, the efficacy of physical activity in patients is unknown. METHODS Eleven genetically diagnosed DM1 patients were recruited to examine the extent to which 12-weeks of cycling can recuperate clinical, and physiological metrics. Furthermore, we studied the underlying molecular mechanisms through which exercise elicits benefits in skeletal muscle of DM1 patients. RESULTS DM1 was associated with impaired muscle function, fitness, and lung capacity. Cycling evoked several clinical, physical, and metabolic advantages in DM1 patients. We highlight that exercise-induced molecular and cellular alterations in patients do not conform with previously published data in murine models and propose a significant role of mitochondrial function in DM1 pathology. Lastly, we discovered a subset of small nucleolar RNAs (snoRNAs) that correlated to indicators of disease severity. CONCLUSION With no available cures, our data supports the efficacy of exercise as a primary intervention to partially mitigate the clinical progression of DM1. Additionally, we provide evidence for the involvement of snoRNAs and other noncoding RNAs in DM1 pathophysiology. TRIAL REGISTRATION This trial was approved by the HiREB committee (#7901) and registered under ClinicalTrials.gov (NCT04187482). FUNDING This work was primarily supported by Neil and Leanne Petroff. This study was also supported by a Canadian Institutes of Health Research Foundation Grant to MAT (#143325).
Collapse
Affiliation(s)
- Andrew I Mikhail
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | - Peter L Nagy
- Department of Neurology, Praxis Genomics, Atlanta, United States of America
| | - Katherine Manta
- Department of Pediatrics, McMaster University Children's Hospital, Hamilton, Canada
| | - Nicholas Rouse
- Department of Neurology, Praxis Genomics, Atlanta, United States of America
| | - Alexander Manta
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | - Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton, Canada
| | - Michael F Nagy
- Department of Neurology, Praxis Genomics, Atlanta, United States of America
| | - Paul Smith
- Department of Neurology, Praxis Genomics, Atlanta, United States of America
| | - Jian-Qiang Lu
- Pathology and Molecular Medicine/Neuropathology, McMaster University, Hamilton, Canada
| | - Joshua P Nederveen
- Department of Pediatrics, McMaster University Children's Hospital, Hamilton, Canada
| | | | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University Children's Hospital, Hamilton, Canada
| |
Collapse
|
26
|
García-Puga M, Saenz-Antoñanzas A, Matheu A, López de Munain A. Targeting Myotonic Dystrophy Type 1 with Metformin. Int J Mol Sci 2022; 23:ijms23052901. [PMID: 35270043 PMCID: PMC8910924 DOI: 10.3390/ijms23052901] [Citation(s) in RCA: 9] [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: 01/24/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disorder of genetic origin. Progressive muscular weakness, atrophy and myotonia are its most prominent neuromuscular features, while additional clinical manifestations in multiple organs are also common. Overall, DM1 features resemble accelerated aging. There is currently no cure or specific treatment for myotonic dystrophy patients. However, in recent years a great effort has been made to identify potential new therapeutic strategies for DM1 patients. Metformin is a biguanide antidiabetic drug, with potential to delay aging at cellular and organismal levels. In DM1, different studies revealed that metformin rescues multiple phenotypes of the disease. This review provides an overview of recent findings describing metformin as a novel therapy to combat DM1 and their link with aging.
Collapse
Affiliation(s)
- Mikel García-Puga
- Neuromuscular Diseases Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED-CIBER), Carlos III Institute, 28031 Madrid, Spain
| | - Ander Saenz-Antoñanzas
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
- Basque Foundation for Science (IKERBASQUE), 48009 Bilbao, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERfes), Carlos III Institute, 28029 Madrid, Spain
- Correspondence: (A.M.); (A.L.d.M.); Tel.: +34-943-006-073 (A.M.); +34-943-006-294 (A.L.d.M.)
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Biodonostia Health Research Institute, 20014 San Sebastian, Spain;
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED-CIBER), Carlos III Institute, 28031 Madrid, Spain
- Neurology Department, Donostia University Hospital, OSAKIDETZA, 20014 San Sebastian, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country, 20014 San Sebastian, Spain
- Correspondence: (A.M.); (A.L.d.M.); Tel.: +34-943-006-073 (A.M.); +34-943-006-294 (A.L.d.M.)
| |
Collapse
|
27
|
Kong HE, Pollack BP. Cutaneous findings in myotonic dystrophy. JAAD Int 2022; 7:7-12. [PMID: 35243403 PMCID: PMC8867117 DOI: 10.1016/j.jdin.2021.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 11/06/2022] Open
Abstract
Myotonic dystrophy types 1 and 2 are a group of complex genetic disorders resulting from the expansion of (CTG)n nucleotide repeats in the DMPK gene. In addition to the hallmark manifestations of myotonia and skeletal muscle atrophy, myotonic dystrophy also affects a myriad of other organs including the heart, lungs, as well as the skin. The most common cutaneous manifestations of myotonic dystrophy are early male frontal alopecia and adult-onset pilomatricomas. Myotonic dystrophy also increases the risk of developing malignant skin diseases such as basal cell carcinoma and melanoma. To aid in the diagnosis and treatment of myotonic dystrophy related skin conditions, it is important for the dermatologist to become cognizant of the common and rare cutaneous manifestations of this genetic disorder. We performed a PubMed search using the key terms “myotonic dystrophy” AND “cutaneous” OR “skin” OR “dermatologic” AND “manifestation” OR “finding.” The resulting publications were manually reviewed for additional relevant publications, and subsequent additional searches were performed as needed, especially regarding the molecular mechanisms of pathogenesis. In this review, we aim to provide an overview of myotonic dystrophy types 1 and 2 and summarize their cutaneous manifestations as well as potential mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Ha Eun Kong
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia
| | - Brian P Pollack
- Atlanta VA Health System, Decatur, Georgia.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute of Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
28
|
Soltanzadeh P. Myotonic Dystrophies: A Genetic Overview. Genes (Basel) 2022; 13:367. [PMID: 35205411 PMCID: PMC8872148 DOI: 10.3390/genes13020367] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophies (DM) are the most common muscular dystrophies in adults, which can affect other non-skeletal muscle organs such as the heart, brain and gastrointestinal system. There are two genetically distinct types of myotonic dystrophy: myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2), both dominantly inherited with significant overlap in clinical manifestations. DM1 results from CTG repeat expansions in the 3'-untranslated region (3'UTR) of the DMPK (dystrophia myotonica protein kinase) gene on chromosome 19, while DM2 is caused by CCTG repeat expansions in intron 1 of the CNBP (cellular nucleic acid-binding protein) gene on chromosome 3. Recent advances in genetics and molecular biology, especially in the field of RNA biology, have allowed better understanding of the potential pathomechanisms involved in DM. In this review article, core clinical features and genetics of DM are presented followed by a discussion on the current postulated pathomechanisms and therapeutic approaches used in DM, including the ones currently in human clinical trial phase.
Collapse
Affiliation(s)
- Payam Soltanzadeh
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Disrupting the Molecular Pathway in Myotonic Dystrophy. Int J Mol Sci 2021; 22:ijms222413225. [PMID: 34948025 PMCID: PMC8708683 DOI: 10.3390/ijms222413225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/26/2023] Open
Abstract
Myotonic dystrophy is the most common muscular dystrophy in adults. It consists of two forms: type 1 (DM1) and type 2 (DM2). DM1 is associated with a trinucleotide repeat expansion mutation, which is transcribed but not translated into protein. The mutant RNA remains in the nucleus, which leads to a series of downstream abnormalities. DM1 is widely considered to be an RNA-based disorder. Thus, we consider three areas of the RNA pathway that may offer targeting opportunities to disrupt the production, stability, and degradation of the mutant RNA.
Collapse
|
30
|
Cerro-Herreros E, González-Martínez I, Moreno N, Espinosa-Espinosa J, Fernández-Costa JM, Colom-Rodrigo A, Overby SJ, Seoane-Miraz D, Poyatos-García J, Vilchez JJ, López de Munain A, Varela MA, Wood MJ, Pérez-Alonso M, Llamusí B, Artero R. Preclinical characterization of antagomiR-218 as a potential treatment for myotonic dystrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:174-191. [PMID: 34513303 PMCID: PMC8413838 DOI: 10.1016/j.omtn.2021.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 07/17/2021] [Indexed: 12/13/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is a rare neuromuscular disease caused by expansion of unstable CTG repeats in a non-coding region of the DMPK gene. CUG expansions in mutant DMPK transcripts sequester MBNL1 proteins in ribonuclear foci. Depletion of this protein is a primary contributor to disease symptoms such as muscle weakness and atrophy and myotonia, yet upregulation of endogenous MBNL1 levels may compensate for this sequestration. Having previously demonstrated that antisense oligonucleotides against miR-218 boost MBNL1 expression and rescue phenotypes in disease models, here we provide preclinical characterization of an antagomiR-218 molecule using the HSALR mouse model and patient-derived myotubes. In HSALR, antagomiR-218 reached 40–60 pM 2 weeks after injection, rescued molecular and functional phenotypes in a dose- and time-dependent manner, and showed a good toxicity profile after a single subcutaneous administration. In muscle tissue, antagomiR rescued the normal subcellular distribution of Mbnl1 and did not alter the proportion of myonuclei containing CUG foci. In patient-derived cells, antagomiR-218 improved defective fusion and differentiation and rescued up to 34% of the gene expression alterations found in the transcriptome of patient cells. Importantly, miR-218 was found to be upregulated in DM1 muscle biopsies, pinpointing this microRNA (miRNA) as a relevant therapeutic target.
Collapse
Affiliation(s)
- Estefanía Cerro-Herreros
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Irene González-Martínez
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Nerea Moreno
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Jorge Espinosa-Espinosa
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Juan M Fernández-Costa
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Anna Colom-Rodrigo
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Sarah J Overby
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - David Seoane-Miraz
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headley Way, OX3 9DU, Oxford, UK.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Javier Poyatos-García
- The IISLAFE Health Research Institute, Avenida Fernando Abril Martorell, 106 Torre A 7 planta, 46026 Valencia, Spain.,Neuromuscular Reference Centre ERN EURO-NMD and Neuromuscular Pathology and Ataxia Research Group, Hospital La Fe Health Research Institute, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan J Vilchez
- The IISLAFE Health Research Institute, Avenida Fernando Abril Martorell, 106 Torre A 7 planta, 46026 Valencia, Spain.,Neuromuscular Reference Centre ERN EURO-NMD and Neuromuscular Pathology and Ataxia Research Group, Hospital La Fe Health Research Institute, Valencia, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Adolfo López de Munain
- Biodonostia Health Research Institute, P° Dr. Beguiristain s/n, 20014 Donostia-San Sebastián, Spain.,Hospital Universitario Donostia-Osakidetza-Departamento de Neurociencias-Universidad del Pais Vasco-CIBERNED
| | - Miguel A Varela
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headley Way, OX3 9DU, Oxford, UK.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Matthew J Wood
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headley Way, OX3 9DU, Oxford, UK.,MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Manuel Pérez-Alonso
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Beatriz Llamusí
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| | - Rubén Artero
- University Research Institute for Biotechnology and Biomedicine (BIOTECMED), Universidad de Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.,Incliva Biomedical Research Institute, Avenida Menéndez Pelayo 4 acc, 46010 Valencia, Spain
| |
Collapse
|
31
|
De Serres-Bérard T, Pierre M, Chahine M, Puymirat J. Deciphering the mechanisms underlying brain alterations and cognitive impairment in congenital myotonic dystrophy. Neurobiol Dis 2021; 160:105532. [PMID: 34655747 DOI: 10.1016/j.nbd.2021.105532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic and heterogeneous disorder caused by the expansion of CTG repeats in the 3' UTR of the myotonic dystrophy protein kinase (DMPK) gene. There is a congenital form (CDM1) of the disease characterized by severe hypotonia, respiratory insufficiency as well as developmental delays and intellectual disabilities. CDM1 infants manifest important brain structure abnormalities present from birth while, in contrast, older patients with adult-onset DM1 often present neurodegenerative features and milder progressive cognitive deficits. Promising therapies targeting central molecular mechanisms contributing to the symptoms of adult-onset DM1 are currently in development, but their relevance for treating cognitive impairment in CDM1, which seems to be a partially distinct neurodevelopmental disorder, remain to be elucidated. Here, we provide an update on the clinical presentation of CDM1 and review recent in vitro and in vivo models that have provided meaningful insights on its consequences in development, with a particular focus on the brain. We discuss how enhanced toxic gain-of-function of the mutated DMPK transcripts with larger CUG repeats and the resulting dysregulation of RNA-binding proteins may affect the developing cortex in utero. Because the methylation of CpG islets flanking the trinucleotide repeats has emerged as a strong biomarker of CDM1, we highlight the need to investigate the tissue-specific impacts of these chromatin modifications in the brain. Finally, we outline promising potential therapeutic treatments for CDM1 and propose future in vitro and in vivo models with great potential to shed light on this disease.
Collapse
Affiliation(s)
- Thiéry De Serres-Bérard
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada; CERVO Brain Research Center, Institut universitaire en santé mentale de Québec, Quebec City, Canada
| | - Marion Pierre
- CERVO Brain Research Center, Institut universitaire en santé mentale de Québec, Quebec City, Canada
| | - Mohamed Chahine
- CERVO Brain Research Center, Institut universitaire en santé mentale de Québec, Quebec City, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada.
| | - Jack Puymirat
- LOEX, CHU de Québec-Université Laval Research Center, Quebec City, Canada; Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
32
|
Liu J, Guo ZN, Yan XL, Yang Y, Huang S. Brain Pathogenesis and Potential Therapeutic Strategies in Myotonic Dystrophy Type 1. Front Aging Neurosci 2021; 13:755392. [PMID: 34867280 PMCID: PMC8634727 DOI: 10.3389/fnagi.2021.755392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy that affects multiple systems including the muscle and heart. The mutant CTG expansion at the 3'-UTR of the DMPK gene causes the expression of toxic RNA that aggregate as nuclear foci. The foci then interfere with RNA-binding proteins, affecting hundreds of mis-spliced effector genes, leading to aberrant alternative splicing and loss of effector gene product functions, ultimately resulting in systemic disorders. In recent years, increasing clinical, imaging, and pathological evidence have indicated that DM1, though to a lesser extent, could also be recognized as true brain diseases, with more and more researchers dedicating to develop novel therapeutic tools dealing with it. In this review, we summarize the current advances in the pathogenesis and pathology of central nervous system (CNS) deficits in DM1, intervention measures currently being investigated are also highlighted, aiming to promote novel and cutting-edge therapeutic investigations.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Shuo Huang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
33
|
Rapisarda A, Bargiela A, Llamusi B, Pont I, Estrada-Tejedor R, Garcia-España E, Artero R, Perez-Alonso M. Defined D-hexapeptides bind CUG repeats and rescue phenotypes of myotonic dystrophy myotubes in a Drosophila model of the disease. Sci Rep 2021; 11:19417. [PMID: 34593893 PMCID: PMC8484449 DOI: 10.1038/s41598-021-98866-0] [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: 12/09/2020] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
In Myotonic Dystrophy type 1 (DM1), a non-coding CTG repeats rare expansion disease; toxic double-stranded RNA hairpins sequester the RNA-binding proteins Muscleblind-like 1 and 2 (MBNL1 and 2) and trigger other DM1-related pathogenesis pathway defects. In this paper, we characterize four d-amino acid hexapeptides identified together with abp1, a peptide previously shown to stabilize CUG RNA in its single-stranded conformation. With the generalized sequence cpy(a/t)(q/w)e, these related peptides improved three MBNL-regulated exon inclusions in DM1-derived cells. Subsequent experiments showed that these compounds generally increased the relative expression of MBNL1 and its nuclear-cytoplasmic distribution, reduced hyperactivated autophagy, and increased the percentage of differentiated (Desmin-positive) cells in vitro. All peptides rescued atrophy of indirect flight muscles in a Drosophila model of the disease, and partially rescued muscle function according to climbing and flight tests. Investigation of their mechanism of action supports that all four compounds can bind to CUG repeats with slightly different association constant, but binding did not strongly influence the secondary structure of the toxic RNA in contrast to abp1. Finally, molecular modeling suggests a detailed view of the interactions of peptide-CUG RNA complexes useful in the chemical optimization of compounds.
Collapse
Affiliation(s)
- Anna Rapisarda
- Department of Genetics, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100, Burjasot, Valencia, Spain.,Translational Genomics Group, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | - Ariadna Bargiela
- Department of Genetics, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100, Burjasot, Valencia, Spain. .,Translational Genomics Group, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain.
| | - Beatriz Llamusi
- Department of Genetics, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100, Burjasot, Valencia, Spain.,Translational Genomics Group, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain.,Arthex Biotech S.L. Catedrático Agustín Escardino 9, Parc Scientific University of Valencia, Paterna, Valencia, Spain
| | - Isabel Pont
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Inorgánica, C. Catedrático José Beltrán 2, Universidad de Valencia, 46980, Paterna, Spain
| | | | - Enrique Garcia-España
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Inorgánica, C. Catedrático José Beltrán 2, Universidad de Valencia, 46980, Paterna, Spain
| | - Ruben Artero
- Department of Genetics, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100, Burjasot, Valencia, Spain.,Translational Genomics Group, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | - Manuel Perez-Alonso
- Department of Genetics, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100, Burjasot, Valencia, Spain.,Translational Genomics Group, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| |
Collapse
|
34
|
Sabater-Arcis M, Bargiela A, Moreno N, Poyatos-Garcia J, Vilchez JJ, Artero R. Musashi-2 contributes to myotonic dystrophy muscle dysfunction by promoting excessive autophagy through miR-7 biogenesis repression. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:652-667. [PMID: 34589284 PMCID: PMC8463325 DOI: 10.1016/j.omtn.2021.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023]
Abstract
Skeletal muscle symptoms strongly contribute to mortality of myotonic dystrophy type 1 (DM1) patients. DM1 is a neuromuscular genetic disease caused by CTG repeat expansions that, upon transcription, sequester the Muscleblind-like family of proteins and dysregulate alternative splicing of hundreds of genes. However, mis-splicing does not satisfactorily explain muscle atrophy and wasting, and several other contributing factors have been suggested, including hyperactivated autophagy leading to excessive catabolism. MicroRNA (miR)-7 has been demonstrated to be necessary and sufficient to repress the autophagy pathway in cell models of the disease, but the origin of its low levels in DM1 was unknown. We have found that the RNA-binding protein Musashi-2 (MSI2) is upregulated in patient-derived myoblasts and biopsy samples. Because it has been previously reported that MSI2 controls miR-7 biogenesis, we tested the hypothesis that excessive MSI2 was repressing miR-7 maturation. Using gene-silencing strategies (small interfering RNAs [siRNAs] and gapmers) and the small molecule MSI2-inhibitor Ro 08-2750, we demonstrate that reducing MSI2 levels or activity boosts miR-7 expression, represses excessive autophagy, and downregulates atrophy-related genes of the UPS system. We also detect a significant upregulation of MBNL1 upon MSI2 silencing. Taken together, we propose MSI2 as a new therapeutic target to treat muscle dysfunction in DM1.
Collapse
Affiliation(s)
- Maria Sabater-Arcis
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Valencia, Spain
- INCLIVA Biomedical Research Institute, 46100 Burjasot, Valencia, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Valencia, Spain
- INCLIVA Biomedical Research Institute, 46100 Burjasot, Valencia, Spain
- Corresponding author: Ariadna Bargiela, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr. Moliner, 50, 46100 Burjasot, Valencia, Spain.
| | - Nerea Moreno
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Valencia, Spain
- INCLIVA Biomedical Research Institute, 46100 Burjasot, Valencia, Spain
| | - Javier Poyatos-Garcia
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
- Neuromuscular Research Unit, Neurology Department, Instituto de Investigación Sanitaria la Fe, Hospital Universitari i Politécnic La Fe, 46026 Valencia, Spain
| | - Juan J. Vilchez
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
- Neuromuscular Research Unit, Neurology Department, Instituto de Investigación Sanitaria la Fe, Hospital Universitari i Politécnic La Fe, 46026 Valencia, Spain
| | - Ruben Artero
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Valencia, Spain
- INCLIVA Biomedical Research Institute, 46100 Burjasot, Valencia, Spain
| |
Collapse
|
35
|
Zelada D, Barrantes FJ, Henríquez JP. Lithium causes differential effects on postsynaptic stability in normal and denervated neuromuscular synapses. Sci Rep 2021; 11:17285. [PMID: 34446751 PMCID: PMC8390761 DOI: 10.1038/s41598-021-96708-7] [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] [Received: 02/13/2021] [Accepted: 08/06/2021] [Indexed: 11/12/2022] Open
Abstract
Lithium chloride has been widely used as a therapeutic mood stabilizer. Although cumulative evidence suggests that lithium plays modulatory effects on postsynaptic receptors, the underlying mechanism by which lithium regulates synaptic transmission has not been fully elucidated. In this work, by using the advantageous neuromuscular synapse, we evaluated the effect of lithium on the stability of postsynaptic nicotinic acetylcholine receptors (nAChRs) in vivo. We found that in normally innervated neuromuscular synapses, lithium chloride significantly decreased the turnover of nAChRs by reducing their internalization. A similar response was observed in CHO-K1/A5 cells expressing the adult muscle-type nAChRs. Strikingly, in denervated neuromuscular synapses, lithium led to enhanced nAChR turnover and density by increasing the incorporation of new nAChRs. Lithium also potentiated the formation of unstable nAChR clusters in non-synaptic regions of denervated muscle fibres. We found that denervation-dependent re-expression of the foetal nAChR γ-subunit was not altered by lithium. However, while denervation inhibits the distribution of β-catenin within endplates, lithium-treated fibres retain β-catenin staining in specific foci of the synaptic region. Collectively, our data reveal that lithium treatment differentially affects the stability of postsynaptic receptors in normal and denervated neuromuscular synapses in vivo, thus providing novel insights into the regulatory effects of lithium on synaptic organization and extending its potential therapeutic use in conditions affecting the peripheral nervous system.
Collapse
Affiliation(s)
- Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, CMA Bio-Bio, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Francisco J Barrantes
- Pontificia Universidad Católica Argentina (UCA)-Scientific and Technological Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, CMA Bio-Bio, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepción, Chile.
| |
Collapse
|
36
|
Demuro S, Di Martino RMC, Ortega JA, Cavalli A. GSK-3β, FYN, and DYRK1A: Master Regulators in Neurodegenerative Pathways. Int J Mol Sci 2021; 22:9098. [PMID: 34445804 PMCID: PMC8396491 DOI: 10.3390/ijms22169098] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Protein kinases (PKs) have been recognized as central nervous system (CNS)-disease-relevant targets due to their master regulatory role in different signal transduction cascades in the neuroscience space. Among them, GSK-3β, FYN, and DYRK1A play a crucial role in the neurodegeneration context, and the deregulation of all three PKs has been linked to different CNS disorders with unmet medical needs, including Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal lobar degeneration (FTLD), and several neuromuscular disorders. The multifactorial nature of these diseases, along with the failure of many advanced CNS clinical trials, and the lengthy approval process of a novel CNS drug have strongly limited the CNS drug discovery. However, in the near-decade from 2010 to 2020, several computer-assisted drug design strategies have been combined with synthetic efforts to develop potent and selective GSK-3β, FYN, and DYRK1A inhibitors as disease-modifying agents. In this review, we described both structural and functional aspects of GSK-3β, FYN, and DYRK1A and their involvement and crosstalk in different CNS pathological signaling pathways. Moreover, we outlined attractive medicinal chemistry approaches including multi-target drug design strategies applied to overcome some limitations of known PKs inhibitors and discover improved modulators with suitable blood-brain barrier (BBB) permeability and drug-like properties.
Collapse
Affiliation(s)
- Stefania Demuro
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Rita M. C. Di Martino
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
| | - Jose A. Ortega
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
| | - Andrea Cavalli
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
37
|
Fernández-Garibay X, Ortega MA, Cerro-Herreros E, Comelles J, Martínez E, Artero R, Fernández-Costa JM, Ramón-Azcón J. Bioengineered in vitro3D model of myotonic dystrophy type 1 human skeletal muscle. Biofabrication 2021; 13. [PMID: 33836519 DOI: 10.1088/1758-5090/abf6ae] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common hereditary myopathy in the adult population. The disease is characterized by progressive skeletal muscle degeneration that produces severe disability. At present, there is still no effective treatment for DM1 patients, but the breakthroughs in understanding the molecular pathogenic mechanisms in DM1 have allowed the testing of new therapeutic strategies. Animal models andin vitrotwo-dimensional cell cultures have been essential for these advances. However, serious concerns exist regarding how faithfully these models reproduce the biological complexity of the disease. Biofabrication tools can be applied to engineer human three-dimensional (3D) culture systems that complement current preclinical research models. Here, we describe the development of the firstin vitro3D model of DM1 human skeletal muscle. Transdifferentiated myoblasts from patient-derived fibroblasts were encapsulated in micromolded gelatin methacryloyl-carboxymethyl cellulose methacrylate hydrogels through photomold patterning on functionalized glass coverslips. These hydrogels present a microstructured topography that promotes myoblasts alignment and differentiation resulting in highly aligned myotubes from both healthy and DM1 cells in a long-lasting cell culture. The DM1 3D microtissues recapitulate the molecular alterations detected in patient biopsies. Importantly, fusion index analyses demonstrate that 3D micropatterning significantly improved DM1 cell differentiation into multinucleated myotubes compared to standard cell cultures. Moreover, the characterization of the 3D cultures of DM1 myotubes detects phenotypes as the reduced thickness of myotubes that can be used for drug testing. Finally, we evaluated the therapeutic effect of antagomiR-23b administration on bioengineered DM1 skeletal muscle microtissues. AntagomiR-23b treatment rescues both molecular DM1 hallmarks and structural phenotype, restoring myotube diameter to healthy control sizes. Overall, these new microtissues represent an improvement over conventional cell culture models and can be used as biomimetic platforms to establish preclinical studies for myotonic dystrophy.
Collapse
Affiliation(s)
- Xiomara Fernández-Garibay
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - María A Ortega
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Estefanía Cerro-Herreros
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Jordi Comelles
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain
| | - Elena Martínez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, E28029 Madrid, Spain
| | - Rubén Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Juan M Fernández-Costa
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Institució Catalana de Reserca I Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010 Barcelona, Spain
| |
Collapse
|
38
|
Yadava RS, Mandal M, Giese JM, Rigo F, Bennett CF, Mahadevan MS. Modeling muscle regeneration in RNA toxicity mice. Hum Mol Genet 2021; 30:1111-1130. [PMID: 33864373 PMCID: PMC8188403 DOI: 10.1093/hmg/ddab108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/04/2023] Open
Abstract
RNA toxicity underlies the pathogenesis of disorders such as myotonic dystrophy type 1 (DM1). Muscular dystrophy is a key element of the pathology of DM1. The means by which RNA toxicity causes muscular dystrophy in DM1 is unclear. Here, we have used the DM200 mouse model of RNA toxicity due to the expression of a mutant DMPK 3′UTR mRNA to model the effects of RNA toxicity on muscle regeneration. Using a BaCl2-induced damage model, we find that RNA toxicity leads to decreased expression of PAX7, and decreased numbers of satellite cells, the stem cells of adult skeletal muscle (also known as MuSCs). This is associated with a delay in regenerative response, a lack of muscle fiber maturation and an inability to maintain a normal number of satellite cells. Repeated muscle damage also elicited key aspects of muscular dystrophy, including fat droplet deposition and increased fibrosis, and the results represent one of the first times to model these classic markers of dystrophic changes in the skeletal muscles of a mouse model of RNA toxicity. Using a ligand-conjugated antisense (LICA) oligonucleotide ASO targeting DMPK sequences for the first time in a mouse model of RNA toxicity in DM1, we find that treatment with IONIS 877864, which targets the DMPK 3′UTR mRNA, is efficacious in correcting the defects in regenerative response and the reductions in satellite cell numbers caused by RNA toxicity. These results demonstrate the possibilities for therapeutic interventions to mitigate the muscular dystrophy associated with RNA toxicity in DM1.
Collapse
Affiliation(s)
- Ramesh S Yadava
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jack M Giese
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA 90210, USA
| | | | - Mani S Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
39
|
Pascual-Gilabert M, López-Castel A, Artero R. Myotonic dystrophy type 1 drug development: A pipeline toward the market. Drug Discov Today 2021; 26:1765-1772. [PMID: 33798646 PMCID: PMC8372527 DOI: 10.1016/j.drudis.2021.03.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/28/2021] [Accepted: 03/23/2021] [Indexed: 01/12/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic neuromuscular genetic disease with an estimated prevalence of approximately at least half a million individuals based on its vast ethnic variation. Building upon a well-known physiopathology and several proof-of-concept therapeutic approaches, herein we compile a comprehensive overview of the most recent drug development programs under preclinical and clinical evaluation. Specifically, close to two dozen drug developments, eight of which are already in clinical trials, explore a diversity of new chemical entities, drug repurposing, oligonucleotide, and gene therapy-based approaches. Of these, repurposing of tideglusib, mexiletine, or metformin appear to be therapies with the most potential to receive marketing authorization for DM1.
Collapse
Affiliation(s)
| | - Arturo López-Castel
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain.
| | - Ruben Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| |
Collapse
|
40
|
Grande V, Hathazi D, O'Connor E, Marteau T, Schara-Schmidt U, Hentschel A, Gourdon G, Nikolenko N, Lochmüller H, Roos A. Dysregulation of GSK3β-Target Proteins in Skin Fibroblasts of Myotonic Dystrophy Type 1 (DM1) Patients. J Neuromuscul Dis 2021; 8:603-619. [PMID: 33682722 DOI: 10.3233/jnd-200558] [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] [Indexed: 12/18/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common monogenetic muscular disorder of adulthood. This multisystemic disease is caused by CTG repeat expansion in the 3'-untranslated region of the DM1 protein kinase gene called DMPK. DMPK encodes a myosin kinase expressed in skeletal muscle cells and other cellular populations such as smooth muscle cells, neurons and fibroblasts. The resultant expanded (CUG)n RNA transcripts sequester RNA binding factors leading to ubiquitous and persistent splicing deregulation. The accumulation of mutant CUG repeats is linked to increased activity of glycogen synthase kinase 3 beta (GSK3β), a highly conserved and ubiquitous serine/threonine kinase with functions in pathways regulating inflammation, metabolism, oncogenesis, neurogenesis and myogenesis. As GSK3β-inhibition ameliorates defects in myogenesis, muscle strength and myotonia in a DM1 mouse model, this kinase represents a key player of DM1 pathobiochemistry and constitutes a promising therapeutic target. To better characterise DM1 patients, and monitor treatment responses, we aimed to define a set of robust disease and severity markers linked to GSK3βby unbiased proteomic profiling utilizing fibroblasts derived from DM1 patients with low (80- 150) and high (2600- 3600) CTG-repeats. Apart from GSK3β increase, we identified dysregulation of nine proteins (CAPN1, CTNNB1, CTPS1, DNMT1, HDAC2, HNRNPH3, MAP2K2, NR3C1, VDAC2) modulated by GSK3β. In silico-based expression studies confirmed expression in neuronal and skeletal muscle cells and revealed a relatively elevated abundance in fibroblasts. The potential impact of each marker in the myopathology of DM1 is discussed based on respective function to inform potential uses as severity markers or for monitoring GSK3β inhibitor treatment responses.
Collapse
Affiliation(s)
- Valentina Grande
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany.,Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Emily O'Connor
- Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Theo Marteau
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Ulrike Schara-Schmidt
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Genevieve Gourdon
- Centre de Recherche en Myologie, Association Institut de Myologie, Sorbonne Université, Inserm UMR 974, Paris, France
| | - Nikoletta Nikolenko
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hanns Lochmüller
- 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.,Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.,Centro Nacional de AnálisisGenómico, Center for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Andreas Roos
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany.,Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
41
|
Tanner MK, Tang Z, Thornton CA. Targeted splice sequencing reveals RNA toxicity and therapeutic response in myotonic dystrophy. Nucleic Acids Res 2021; 49:2240-2254. [PMID: 33503262 PMCID: PMC7913682 DOI: 10.1093/nar/gkab022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Biomarker-driven trials hold promise for therapeutic development in chronic diseases, such as muscular dystrophy. Myotonic dystrophy type 1 (DM1) involves RNA toxicity, where transcripts containing expanded CUG-repeats (CUGexp) accumulate in nuclear foci and sequester splicing factors in the Muscleblind-like (Mbnl) family. Oligonucleotide therapies to mitigate RNA toxicity have emerged but reliable measures of target engagement are needed. Here we examined muscle transcriptomes in mouse models of DM1 and found that CUGexp expression or Mbnl gene deletion cause similar dysregulation of alternative splicing. We selected 35 dysregulated exons for further study by targeted RNA sequencing. Across a spectrum of mouse models, the individual splice events and a composite index derived from all events showed a graded response to decrements of Mbnl or increments of CUGexp. Antisense oligonucleotides caused prompt reduction of CUGexp RNA and parallel correction of the splicing index, followed by subsequent elimination of myotonia. These results suggest that targeted splice sequencing may provide a sensitive and reliable way to assess therapeutic impact in DM1.
Collapse
Affiliation(s)
- Matthew K Tanner
- Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Zhenzhi Tang
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Charles A Thornton
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
42
|
Johnson SJ, Cooper TA. Overlapping mechanisms of lncRNA and expanded microsatellite RNA. WILEY INTERDISCIPLINARY REVIEWS. RNA 2021; 12:e1634. [PMID: 33191580 PMCID: PMC7880542 DOI: 10.1002/wrna.1634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022]
Abstract
RNA has major regulatory roles in a wide range of biological processes and a surge of RNA research has led to the classification of numerous functional RNA species. One example is long noncoding RNAs (lncRNAs) that are structurally complex transcripts >200 nucleotides (nt) in length and lacking a canonical open reading frame (ORF). Despite a general lack of sequence conservation and low expression levels, many lncRNAs have been shown to have functionality in diverse biological processes as well as in mechanisms of disease. In parallel with the growing understanding of lncRNA functions, there is a growing subset of microsatellite expansion disorders in which the primary mechanism of pathogenesis is an RNA gain of function arising from RNA transcripts from the mutant allele. Microsatellite expansion disorders are caused by an expansion of short (3-10 nt) repeats located within coding genes. Expanded repeat-containing RNA mediates toxicity through multiple mechanisms, the details of which remain only partially understood. The purpose of this review is to highlight the links between functional mechanisms of lncRNAs and the potential pathogenic mechanisms of expanded microsatellite RNA. These shared mechanisms include protein sequestration, peptide translation, micro-RNA (miRNA) processing, and miRNA sequestration. Recognizing the parallels between the normal functions of lncRNAs and the negative impact of expanded microsatellite RNA on biological processes can provide reciprocal understanding to the roles of both RNA species. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Sara J Johnson
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas A Cooper
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
43
|
Ozimski LL, Sabater-Arcis M, Bargiela A, Artero R. The hallmarks of myotonic dystrophy type 1 muscle dysfunction. Biol Rev Camb Philos Soc 2020; 96:716-730. [PMID: 33269537 DOI: 10.1111/brv.12674] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is the most prevalent form of muscular dystrophy in adults and yet there are currently no treatment options. Although this disease causes multisystemic symptoms, it is mainly characterised by myopathy or diseased muscles, which includes muscle weakness, atrophy, and myotonia, severely affecting the lives of patients worldwide. On a molecular level, DM1 is caused by an expansion of CTG repeats in the 3' untranslated region (3'UTR) of the DM1 Protein Kinase (DMPK) gene which become pathogenic when transcribed into RNA forming ribonuclear foci comprised of auto complementary CUG hairpin structures that can bind proteins. This leads to the sequestration of the muscleblind-like (MBNL) family of proteins, depleting them, and the abnormal stabilisation of CUGBP Elav-like family member 1 (CELF1), enhancing it. Traditionally, DM1 research has focused on this RNA toxicity and how it alters MBNL and CELF1 functions as key splicing regulators. However, other proteins are affected by the toxic DMPK RNA and there is strong evidence that supports various signalling cascades playing an important role in DM1 pathogenesis. Specifically, the impairment of protein kinase B (AKT) signalling in DM1 increases autophagy, apoptosis, and ubiquitin-proteasome activity, which may also be affected in DM1 by AMP-activated protein kinase (AMPK) downregulation. AKT also regulates CELF1 directly, by affecting its subcellular localisation, and indirectly as it inhibits glycogen synthase kinase 3 beta (GSK3β), which stabilises the repressive form of CELF1 in DM1. Another kinase that contributes to CELF1 mis-regulation, in this case by hyperphosphorylation, is protein kinase C (PKC). Additionally, it has been demonstrated that fibroblast growth factor-inducible 14 (Fn14) is induced in DM1 and is associated with downstream signalling through the nuclear factor κB (NFκB) pathways, associating inflammation with this disease. Furthermore, MBNL1 and CELF1 play a role in cytoplasmic processes involved in DM1 myopathy, altering proteostasis and sarcomere structure. Finally, there are many other elements that could contribute to the muscular phenotype in DM1 such as alterations to satellite cells, non-coding RNA metabolism, calcium dysregulation, and repeat-associated non-ATG (RAN) translation. This review aims to organise the currently dispersed knowledge on the different pathways affected in DM1 and discusses the unexplored connections that could potentially help in providing new therapeutic targets in DM1 research.
Collapse
Affiliation(s)
- Lauren L Ozimski
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain.,Arthex Biotech, Catedrático Escardino, 9, Paterna, Valencia, 46980, Spain
| | - Maria Sabater-Arcis
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| | - Ruben Artero
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menéndez Pelayo 4 acc., Valencia, 46010, Spain.,University Institute for Biotechnology and Biomedicine, Dr. Moliner 50, Burjasot, Valencia, 46100, Spain.,CIPF-INCLIVA Joint Unit, Valencia, 46012, Spain
| |
Collapse
|
44
|
Horrigan J, Gomes TB, Snape M, Nikolenko N, McMorn A, Evans S, Yaroshinsky A, Della Pasqua O, Oosterholt S, Lochmüller H. A Phase 2 Study of AMO-02 (Tideglusib) in Congenital and Childhood-Onset Myotonic Dystrophy Type 1 (DM1). Pediatr Neurol 2020; 112:84-93. [PMID: 32942085 DOI: 10.1016/j.pediatrneurol.2020.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND GSK3β is an intracellular regulatory kinase that is dysregulated in multiple tissues in type 1 myotonic dystrophy, a rare neuromuscular disorder that manifests at any age. AMO-02 (tideglusib) inhibits GSK3β activity in preclinical models of type 1 myotonic dystrophy and promotes cellular maturation as well as normalizes aberrant molecular and behavioral phenotypes. This phase 2 study assessed the pharmacokinetics, safety and tolerability, and preliminary efficacy of AMO-02 in adolescents and adults with congenital and childhood-onset type 1 myotonic dystrophy. METHODS Sixteen subjects (aged 13 to 34 years) with congenital and childhood-onset type 1 myotonic dystrophy received 12 weeks of single-blind fixed-dose oral treatment with either 400 mg (n = 8) or 1000 mg (n = 8) AMO-02 (NCT02858908). Blood samples were obtained for pharmacokinetic assessment. Safety assessments, such as laboratory tests and electrocardiograms, as well as efficacy assessments of syndromal, cognitive, and muscular functioning, were obtained. RESULTS AMO-02 plasma concentrations conformed to a two-compartment model with first-order absorption and elimination, and dose-dependent increases in exposure (area under the curve) were observed. AMO-02 was generally safe and well-tolerated. No early discontinuations due to adverse events or dose adjustments of AMO-02 occurred. The majority of subjects manifested clinical improvement in their central nervous system and neuromuscular symptoms after 12 weeks of treatment compared with the placebo baseline, with a larger response noted at the 1000 mg/day dose level. AMO-02 exposure (cumulative area under the curve) was significantly correlated (P < 0.01) with change from baseline on several key efficacy assessments. CONCLUSION AMO-02 has favorable pharmacokinetic and clinical risk/benefit profiles meriting further study as a potential treatment for congenital and childhood-onset type 1 myotonic dystrophy.
Collapse
Affiliation(s)
| | - Tiago Bernardino Gomes
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, UK
| | | | - Nikoletta Nikolenko
- National Hospital for Neurology and Neurosurgery, Queen Square, University College London Hospitals NHS Foundation Trust, London, UK
| | | | | | | | - Oscar Della Pasqua
- Clinical Pharmacology and Therapeutics, University College London, London, UK
| | - Sean Oosterholt
- Clinical Pharmacology and Therapeutics, University College London, London, UK
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute; Division of Neurology, Department of Medicine, The Ottawa Hospital; and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| |
Collapse
|
45
|
Crawford Parks TE, Marcellus KA, Péladeau C, Jasmin BJ, Ravel-Chapuis A. Overexpression of Staufen1 in DM1 mouse skeletal muscle exacerbates dystrophic and atrophic features. Hum Mol Genet 2020; 29:2185-2199. [PMID: 32504084 DOI: 10.1093/hmg/ddaa111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/15/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
In myotonic dystrophy type 1 (DM1), the CUG expansion (CUGexp) in the 3' untranslated region of the dystrophia myotonica protein kinase messenger ribonucleic acid affects the homeostasis of ribonucleic acid-binding proteins, causing the multiple symptoms of DM1. We have previously reported that Staufen1 is increased in skeletal muscles from DM1 mice and patients and that sustained Staufen1 expression in mature mouse muscle causes a progressive myopathy. Here, we hypothesized that the elevated levels of Staufen1 contributes to the myopathic features of the disease. Interestingly, the classic DM1 mouse model human skeletal actin long repeat (HSALR) lacks overt atrophy while expressing CUGexp transcripts and elevated levels of endogenous Staufen1, suggesting a lower sensitivity to atrophic signaling in this model. We report that further overexpression of Staufen1 in the DM1 mouse model HSALR causes a myopathy via inhibition of protein kinase B signaling through an increase in phosphatase tensin homolog, leading to the expression of atrogenes. Interestingly, we also show that Staufen1 regulates the expression of muscleblind-like splicing regulator 1 and CUG-binding protein elav-like family member 1 in wild-type and DM1 skeletal muscle. Together, data obtained from these new DM1 mouse models provide evidence for the role of Staufen1 as an atrophy-associated gene that impacts progressive muscle wasting in DM1. Accordingly, our findings highlight the potential of Staufen1 as a therapeutic target and biomarker.
Collapse
Affiliation(s)
- Tara E Crawford Parks
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Kristen A Marcellus
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Christine Péladeau
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
46
|
Whitley KC, Hamstra SI, Baranowski RW, Watson CJF, MacPherson REK, MacNeil AJ, Roy BD, Vandenboom R, Fajardo VA. GSK3 inhibition with low dose lithium supplementation augments murine muscle fatigue resistance and specific force production. Physiol Rep 2020; 8:e14517. [PMID: 32729236 PMCID: PMC7390913 DOI: 10.14814/phy2.14517] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022] Open
Abstract
Calcineurin is a Ca2+ -dependent serine/threonine phosphatase that dephosphorylates nuclear factor of activated T cells (NFAT), allowing for NFAT entry into the nucleus. In skeletal muscle, calcineurin signaling and NFAT activation increases the expression of proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) and slow myosin heavy chain (MHC) I ultimately promoting fatigue resistance. Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase that antagonizes calcineurin by re-phosphorylating NFAT preventing its entry into the nucleus. Here, we tested whether GSK3 inhibition in vivo with low dose lithium chloride (LiCl) supplementation (10 mg kg-1 day-1 for 6 weeks) in male C57BL/6J mice would enhance muscle fatigue resistance in soleus and extensor digitorum longus (EDL) muscles by activating NFAT and augmenting PGC-1α and MHC I expression. LiCl treatment inhibited GSK3 by elevating Ser9 phosphorylation in soleus (+1.8-fold, p = .007) and EDL (+1.3-fold p = .04) muscles. This was associated with a significant reduction in NFAT phosphorylation (-50%, p = .04) and a significant increase in PGC-1α (+1.5-fold, p = .05) in the soleus but not the EDL. MHC isoform analyses in the soleus also revealed a 1.2-fold increase in MHC I (p = .04) with no change in MHC IIa. In turn, a significant enhancement in soleus muscle fatigue (p = .04), but not EDL (p = .26) was found with LiCl supplementation. Lastly, LiCl enhanced specific force production in both soleus (p < .0001) and EDL (p = .002) muscles. Altogether, our findings show the skleletal muscle contractile benefits of LiCl-mediated GSK3 inhibition in mice.
Collapse
Affiliation(s)
- Kennedy C. Whitley
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| | - Sophie I. Hamstra
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| | - Ryan W. Baranowski
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| | | | | | - Adam J. MacNeil
- Department of Health SciencesBrock UniversitySt. CatharinesONCanada
| | - Brian D. Roy
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| | - Rene Vandenboom
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| | - Val A. Fajardo
- Department of KinesiologyBrock UniversitySt. CatharinesONCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesONCanada
| |
Collapse
|
47
|
Yadava RS, Yu Q, Mandal M, Rigo F, Bennett CF, Mahadevan MS. Systemic therapy in an RNA toxicity mouse model with an antisense oligonucleotide therapy targeting a non-CUG sequence within the DMPK 3'UTR RNA. Hum Mol Genet 2020; 29:1440-1453. [PMID: 32242217 PMCID: PMC7268549 DOI: 10.1093/hmg/ddaa060] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/19/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most common adult muscular dystrophy, is an autosomal dominant disorder caused by an expansion of a (CTG)n tract within the 3' untranslated region (3'UTR) of the dystrophia myotonica protein kinase (DMPK) gene. Mutant DMPK mRNAs are toxic, present in nuclear RNA foci and correlated with a plethora of RNA splicing defects. Cardinal features of DM1 are myotonia and cardiac conduction abnormalities. Using transgenic mice, we have demonstrated that expression of the mutant DMPK 3'UTR is sufficient to elicit these features of DM1. Here, using these mice, we present a study of systemic treatment with an antisense oligonucleotide (ASO) (ISIS 486178) targeted to a non-CUG sequence within the 3'UTR of DMPK. RNA foci and DMPK 3'UTR mRNA levels were reduced in both the heart and skeletal muscles. This correlated with improvements in several splicing defects in skeletal and cardiac muscles. The treatment reduced myotonia and this correlated with increased Clcn1 expression. Furthermore, functional testing showed improvements in treadmill running. Of note, we demonstrate that the ASO treatment reversed the cardiac conduction abnormalities, and this correlated with restoration of Gja5 (connexin 40) expression in the heart. This is the first time that an ASO targeting a non-CUG sequence within the DMPK 3'UTR has demonstrated benefit on the key DM1 phenotypes of myotonia and cardiac conduction defects. Our data also shows for the first time that ASOs may be a viable option for treating cardiac pathology in DM1.
Collapse
Affiliation(s)
- Ramesh S Yadava
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA 90210, USA
| | | | - Mani S Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
48
|
Timchenko L. Correction of RNA-Binding Protein CUGBP1 and GSK3β Signaling as Therapeutic Approach for Congenital and Adult Myotonic Dystrophy Type 1. Int J Mol Sci 2019; 21:ijms21010094. [PMID: 31877772 PMCID: PMC6982105 DOI: 10.3390/ijms21010094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a complex genetic disease affecting many tissues. DM1 is caused by an expansion of CTG repeats in the 3′-UTR of the DMPK gene. The mechanistic studies of DM1 suggested that DMPK mRNA, containing expanded CUG repeats, is a major therapeutic target in DM1. Therefore, the removal of the toxic RNA became a primary focus of the therapeutic development in DM1 during the last decade. However, a cure for this devastating disease has not been found. Whereas the degradation of toxic RNA remains a preferential approach for the reduction of DM1 pathology, other approaches targeting early toxic events downstream of the mutant RNA could be also considered. In this review, we discuss the beneficial role of the restoring of the RNA-binding protein, CUGBP1/CELF1, in the correction of DM1 pathology. It has been recently found that the normalization of CUGBP1 activity with the inhibitors of GSK3 has a positive effect on the reduction of skeletal muscle and CNS pathologies in DM1 mouse models. Surprisingly, the inhibitor of GSK3, tideglusib also reduced the toxic CUG-containing RNA. Thus, the development of the therapeutics, based on the correction of the GSK3β-CUGBP1 pathway, is a promising option for this complex disease.
Collapse
Affiliation(s)
- Lubov Timchenko
- Departments of Neurology and Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
49
|
Increased Muscleblind levels by chloroquine treatment improve myotonic dystrophy type 1 phenotypes in in vitro and in vivo models. Proc Natl Acad Sci U S A 2019; 116:25203-25213. [PMID: 31754023 DOI: 10.1073/pnas.1820297116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a life-threatening and chronically debilitating neuromuscular disease caused by the expansion of a CTG trinucleotide repeat in the 3' UTR of the DMPK gene. The mutant RNA forms insoluble structures capable of sequestering RNA binding proteins of the Muscleblind-like (MBNL) family, which ultimately leads to phenotypes. In this work, we demonstrate that treatment with the antiautophagic drug chloroquine was sufficient to up-regulate MBNL1 and 2 proteins in Drosophila and mouse (HSALR) models and patient-derived myoblasts. Extra Muscleblind was functional at the molecular level and improved splicing events regulated by MBNLs in all disease models. In vivo, chloroquine restored locomotion, rescued average cross-sectional muscle area, and extended median survival in DM1 flies. In HSALR mice, the drug restored muscular strength and histopathology signs and reduced the grade of myotonia. Taken together, these results offer a means to replenish critically low MBNL levels in DM1.
Collapse
|
50
|
Sabater-Arcis M, Bargiela A, Furling D, Artero R. miR-7 Restores Phenotypes in Myotonic Dystrophy Muscle Cells by Repressing Hyperactivated Autophagy. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:278-292. [PMID: 31855836 PMCID: PMC6926285 DOI: 10.1016/j.omtn.2019.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Abstract
Unstable CTG expansions in the 3’ UTR of the DMPK gene are responsible for myotonic dystrophy type 1 (DM1) condition. Muscle dysfunction is one of the main contributors to DM1 mortality and morbidity. Pathways by which mutant DMPK trigger muscle defects, however, are not fully understood. We previously reported that miR-7 was downregulated in a DM1 Drosophila model and in biopsies from patients. Here, using DM1 and normal muscle cells, we investigated whether miR-7 contributes to the muscle phenotype by studying the consequences of replenishing or blocking miR-7, respectively. Restoration of miR-7 with agomiR-7 was sufficient to rescue DM1 myoblast fusion defects and myotube growth. Conversely, oligonucleotide-mediated blocking of miR-7 in normal myoblasts led to fusion and myotube growth defects. miR-7 was found to regulate autophagy and the ubiquitin-proteasome system in human muscle cells. Thus, low levels of miR-7 promoted both processes, and high levels of miR-7 repressed them. Furthermore, we uncovered that the mechanism by which miR-7 improves atrophy-related phenotypes is independent of MBNL1, thus suggesting that miR-7 acts downstream or in parallel to MBNL1. Collectively, these results highlight an unknown function for miR-7 in muscle dysfunction through autophagy- and atrophy-related pathways and support that restoration of miR-7 levels is a candidate therapeutic target for counteracting muscle dysfunction in DM1.
Collapse
Affiliation(s)
- Maria Sabater-Arcis
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain.
| | - Denis Furling
- Sorbonne Université, Inserm, Association Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Ruben Artero
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain
| |
Collapse
|