51
|
López Castel A, Overby SJ, Artero R. MicroRNA-Based Therapeutic Perspectives in Myotonic Dystrophy. Int J Mol Sci 2019; 20:ijms20225600. [PMID: 31717488 PMCID: PMC6888406 DOI: 10.3390/ijms20225600] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Myotonic dystrophy involves two types of chronically debilitating rare neuromuscular diseases: type 1 (DM1) and type 2 (DM2). Both share similarities in molecular cause, clinical signs, and symptoms with DM2 patients usually displaying milder phenotypes. It is well documented that key clinical symptoms in DM are associated with a strong mis-regulation of RNA metabolism observed in patient’s cells. This mis-regulation is triggered by two leading DM-linked events: the sequestration of Muscleblind-like proteins (MBNL) and the mis-regulation of the CUGBP RNA-Binding Protein Elav-Like Family Member 1 (CELF1) that cause significant alterations to their important functions in RNA processing. It has been suggested that DM1 may be treatable through endogenous modulation of the expression of MBNL and CELF1 proteins. In this study, we analyzed the recent identification of the involvement of microRNA (miRNA) molecules in DM and focus on the modulation of these miRNAs to therapeutically restore normal MBNL or CELF1 function. We also discuss additional prospective miRNA targets, the use of miRNAs as disease biomarkers, and additional promising miRNA-based and miRNA-targeting drug development strategies. This review provides a unifying overview of the dispersed data on miRNA available in the context of DM.
Collapse
Affiliation(s)
- Arturo López Castel
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
- Correspondence: (A.L.C.); (R.A.)
| | - Sarah Joann Overby
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
| | - Rubén Artero
- Translational Genomics Group, Incliva Health Research Institute, Burjassot, 46100 Valencia, Spain
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (Eri Biotecmed), University of Valencia, Burjassot, 46100 Valencia, Spain
- Correspondence: (A.L.C.); (R.A.)
| |
Collapse
|
52
|
Kurgan N, Whitley KC, Maddalena LA, Moradi F, Stoikos J, Hamstra SI, Rubie EA, Kumar M, Roy BD, Woodgett JR, Stuart JA, Fajardo VA. A Low-Therapeutic Dose of Lithium Inhibits GSK3 and Enhances Myoblast Fusion in C2C12 Cells. Cells 2019; 8:cells8111340. [PMID: 31671858 PMCID: PMC6912290 DOI: 10.3390/cells8111340] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) slows myogenic differentiation and myoblast fusion partly by inhibiting the Wnt/β-catenin signaling pathway. Lithium, a common medication for bipolar disorder, inhibits GSK3 via Mg+ competition and increased Ser21 (GSK3α) or Ser9 (GSK3β) phosphorylation, leading to enhanced myoblast fusion and myogenic differentiation. However, previous studies demonstrating the effect of lithium on GSK3 have used concentrations up to 10 mM, which greatly exceeds concentrations measured in the serum of patients being treated for bipolar disorder (0.5–1.2 mM). Here, we determined whether a low-therapeutic (0.5 mM) dose of lithium could promote myoblast fusion and myogenic differentiation in C2C12 cells. C2C12 myotubes differentiated for three days in media containing 0.5 mM lithium chloride (LiCl) had significantly higher GSK3β (ser9) and GSK3α (ser21) phosphorylation compared with control myotubes differentiated in the same media without LiCl (+2–2.5 fold, p < 0.05), a result associated with an increase in total β-catenin. To further demonstrate that 0.5 mM LiCl inhibited GSK3 activity, we also developed a novel GSK3-specific activity assay. Using this enzyme-linked spectrophotometric assay, we showed that 0.5 mM LiCl-treated myotubes had significantly reduced GSK3 activity (−86%, p < 0.001). Correspondingly, 0.5 mM LiCl treated myotubes had a higher myoblast fusion index compared with control (p < 0.001) and significantly higher levels of markers of myogenesis (myogenin, +3-fold, p < 0.001) and myogenic differentiation (myosin heavy chain, +10-fold, p < 0.001). These results indicate that a low-therapeutic dose of LiCl is sufficient to promote myoblast fusion and myogenic differentiation in muscle cells, which has implications for the treatment of several myopathic conditions.
Collapse
Affiliation(s)
- Nigel Kurgan
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
- Centre for Bone and Muscle Health, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
| | - Kennedy C. Whitley
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
- Centre for Bone and Muscle Health, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
| | - Lucas A. Maddalena
- Department of Biological Sciences, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (L.A.M.); (F.M.); (J.A.S.)
| | - Fereshteh Moradi
- Department of Biological Sciences, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (L.A.M.); (F.M.); (J.A.S.)
| | - Joshua Stoikos
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
| | - Sophie I. Hamstra
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
- Centre for Bone and Muscle Health, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
| | - Elizabeth A. Rubie
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON M5G 1X5, Canada; (E.A.R.); (M.K.); (J.R.W.)
| | - Megha Kumar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON M5G 1X5, Canada; (E.A.R.); (M.K.); (J.R.W.)
| | - Brian D. Roy
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
- Centre for Bone and Muscle Health, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
| | - James R. Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON M5G 1X5, Canada; (E.A.R.); (M.K.); (J.R.W.)
| | - Jeffrey A. Stuart
- Department of Biological Sciences, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (L.A.M.); (F.M.); (J.A.S.)
| | - Val A. Fajardo
- Department of Kinesiology, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada; (N.K.); (K.C.W.); (J.S.); (S.I.H.); (B.D.R.)
- Centre for Bone and Muscle Health, Brock University 1812 Sir Isaac Brock Way, St. Catharines, ON L2S 3A1, Canada
- Correspondence:
| |
Collapse
|
53
|
Correction of Glycogen Synthase Kinase 3β in Myotonic Dystrophy 1 Reduces the Mutant RNA and Improves Postnatal Survival of DMSXL Mice. Mol Cell Biol 2019; 39:MCB.00155-19. [PMID: 31383751 DOI: 10.1128/mcb.00155-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/01/2019] [Indexed: 11/20/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystem neuromuscular disease without cure. One of the possible therapeutic approaches for DM1 is correction of the RNA-binding proteins CUGBP1 and MBNL1, misregulated in DM1. CUGBP1 activity is controlled by glycogen synthase kinase 3β (GSK3β), which is elevated in skeletal muscle of patients with DM1, and inhibitors of GSK3 were suggested as therapeutic molecules to correct CUGBP1 activity in DM1. Here, we describe that correction of GSK3β with a small-molecule inhibitor of GSK3, tideglusib (TG), not only normalizes the GSK3β-CUGBP1 pathway but also reduces the mutant DMPK mRNA in myoblasts from patients with adult DM1 and congenital DM1 (CDM1). Correction of GSK3β in a mouse model of DM1 (HSALR mice) with TG also reduces the levels of CUG-containing RNA, normalizing a number of CUGBP1- and MBNL1-regulated mRNA targets. We also found that the GSK3β-CUGBP1 pathway is abnormal in skeletal muscle and brain of DMSXL mice, expressing more than 1,000 CUG repeats, and that the correction of this pathway with TG increases postnatal survival and improves growth and neuromotor activity of DMSXL mice. These findings show that the inhibitors of GSK3, such as TG, may correct pathology in DM1 and CDM1 via several pathways.
Collapse
|
54
|
Reddy K, Jenquin JR, Cleary JD, Berglund JA. Mitigating RNA Toxicity in Myotonic Dystrophy using Small Molecules. Int J Mol Sci 2019; 20:E4017. [PMID: 31426500 PMCID: PMC6720693 DOI: 10.3390/ijms20164017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 12/26/2022] Open
Abstract
This review, one in a series on myotonic dystrophy (DM), is focused on the development and potential use of small molecules as therapeutics for DM. The complex mechanisms and pathogenesis of DM are covered in the associated reviews. Here, we examine the various small molecule approaches taken to target the DNA, RNA, and proteins that contribute to disease onset and progression in myotonic dystrophy type 1 (DM1) and 2 (DM2).
Collapse
Affiliation(s)
- Kaalak Reddy
- The RNA Institute, University at Albany-SUNY, Albany, NY 12222, USA.
| | - Jana R Jenquin
- Center for NeuroGenetics and Biochemistry & Molecular Biology, University of Florida, Gainesville, FL 32608, USA
| | - John D Cleary
- The RNA Institute, University at Albany-SUNY, Albany, NY 12222, USA
| | - J Andrew Berglund
- The RNA Institute, University at Albany-SUNY, Albany, NY 12222, USA.
- Center for NeuroGenetics and Biochemistry & Molecular Biology, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
55
|
Carrell ST, Tang Z, Mohr S, Lambowitz AM, Thornton CA. Detection of expanded RNA repeats using thermostable group II intron reverse transcriptase. Nucleic Acids Res 2019; 46:e1. [PMID: 29036654 PMCID: PMC5758912 DOI: 10.1093/nar/gkx867] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/27/2017] [Indexed: 12/15/2022] Open
Abstract
Cellular accumulation of repetitive RNA occurs in several dominantly-inherited genetic disorders. Expanded CUG, CCUG or GGGGCC repeats are expressed in myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), or familial amyotrophic lateral sclerosis, respectively. Expanded repeat RNAs (ER-RNAs) exert a toxic gain-of-function and are prime therapeutic targets in these diseases. However, efforts to quantify ER-RNA levels or monitor knockdown are confounded by stable structure and heterogeneity of the ER-RNA tract and background signal from non-expanded repeats. Here, we used a thermostable group II intron reverse transcriptase (TGIRT-III) to convert ER-RNA to cDNA, followed by quantification on slot blots. We found that TGIRT-III was capable of reverse transcription (RTn) on enzymatically synthesized ER-RNAs. By using conditions that limit cDNA synthesis from off-target sequences, we observed hybridization signals on cDNA slot blots from DM1 and DM2 muscle samples but not from healthy controls. In transgenic mouse models of DM1 the cDNA slot blots accurately reflected the differences of ER-RNA expression across different transgenic lines, and showed therapeutic reductions in skeletal and cardiac muscle, accompanied by improvements of the DM1-associated splicing defects. TGIRT-III was also active on CCCCGG- and GGGGCC-repeats, suggesting that ER-RNA analysis is feasible for several repeat expansion disorders.
Collapse
Affiliation(s)
- Samuel T Carrell
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14611, USA
| | - Zhenzhi Tang
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14611, USA
| | - Sabine Mohr
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Alan M Lambowitz
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Charles A Thornton
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14611, USA
| |
Collapse
|
56
|
Wang ET, Treacy D, Eichinger K, Struck A, Estabrook J, Olafson H, Wang TT, Bhatt K, Westbrook T, Sedehizadeh S, Ward A, Day J, Brook D, Berglund JA, Cooper T, Housman D, Thornton C, Burge C. Transcriptome alterations in myotonic dystrophy skeletal muscle and heart. Hum Mol Genet 2019; 28:1312-1321. [PMID: 30561649 DOI: 10.1093/hmg/ddy432] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 11/12/2022] Open
Abstract
Myotonic dystrophy (dystrophia myotonica, DM) is a multi-systemic disease caused by expanded CTG or CCTG microsatellite repeats. Characterized by symptoms in muscle, heart and central nervous system, among others, it is one of the most variable diseases known. A major pathogenic event in DM is the sequestration of muscleblind-like proteins by CUG or CCUG repeat-containing RNAs transcribed from expanded repeats, and differences in the extent of MBNL sequestration dependent on repeat length and expression level may account for some portion of the variability. However, many other cellular pathways are reported to be perturbed in DM, and the severity of specific disease symptoms varies among individuals. To help understand this variability and facilitate research into DM, we generated 120 RNASeq transcriptomes from skeletal and heart muscle derived from healthy and DM1 biopsies and autopsies. A limited number of DM2 and Duchenne muscular dystrophy samples were also sequenced. We analyzed splicing and gene expression, identified tissue-specific changes in RNA processing and uncovered transcriptome changes strongly correlating with muscle strength. We created a web resource at http://DMseq.org that hosts raw and processed transcriptome data and provides a lightweight, responsive interface that enables browsing of processed data across the genome.
Collapse
Affiliation(s)
- Eric T Wang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL, USA.,Center for NeuroGenetics, University of Florida, Gainesville, FL, USA.,University of Florida Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Daniel Treacy
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katy Eichinger
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | - Adam Struck
- Department of Biochemistry, University of Oregon, Eugene, OR, USA
| | - Joseph Estabrook
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL, USA.,Center for NeuroGenetics, University of Florida, Gainesville, FL, USA.,University of Florida Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Hailey Olafson
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL, USA.,Center for NeuroGenetics, University of Florida, Gainesville, FL, USA.,University of Florida Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Thomas T Wang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kirti Bhatt
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | - Tony Westbrook
- School of Life Sciences, Queen's Medical Center, University of Nottingham, Nottingham, UK
| | - Sam Sedehizadeh
- School of Life Sciences, Queen's Medical Center, University of Nottingham, Nottingham, UK
| | - Amanda Ward
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - John Day
- Department of Neurology, Stanford University, Palo Alto, CA, USA
| | - David Brook
- School of Life Sciences, Queen's Medical Center, University of Nottingham, Nottingham, UK
| | - J Andrew Berglund
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL, USA.,Center for NeuroGenetics, University of Florida, Gainesville, FL, USA.,University of Florida Genetics Institute, University of Florida, Gainesville, FL, USA.,Department of Biochemistry, University of Oregon, Eugene, OR, USA
| | - Thomas Cooper
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David Housman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles Thornton
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | - Christopher Burge
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
57
|
Ravel-Chapuis A, Al-Rewashdy A, Bélanger G, Jasmin BJ. Pharmacological and physiological activation of AMPK improves the spliceopathy in DM1 mouse muscles. Hum Mol Genet 2019; 27:3361-3376. [PMID: 29982462 DOI: 10.1093/hmg/ddy245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/28/2018] [Indexed: 12/26/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a debilitating multisystemic disorder caused by a triplet repeat expansion in the 3' untranslated region of dystrophia myotonica protein kinase mRNAs. Mutant mRNAs accumulate in the nucleus of affected cells and misregulate RNA-binding proteins, thereby promoting characteristic missplicing events. However, little is known about the signaling pathways that may be affected in DM1. Here, we investigated the status of activated protein kinase (AMPK) signaling in DM1 skeletal muscle and found that the AMPK pathway is markedly repressed in a DM1 mouse model (human skeletal actin-long repeat, HSALR) and patient-derived DM1 myoblasts. Chronic pharmacological activation of AMPK signaling in DM1 mice with 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) has multiple beneficial effects on the DM1 phenotype. Indeed, a 6-week AICAR treatment of DM1 mice promoted expression of a slower, more oxidative phenotype, improved muscle histology and corrected several events associated with RNA toxicity. Importantly, AICAR also had a dose-dependent positive effect on the spliceopathy in patient-derived DM1 myoblasts. In separate experiments, we also show that chronic treatment of DM1 mice with resveratrol as well as voluntary wheel running also rescued missplicing events in muscle. Collectively, our findings demonstrate the therapeutic potential of chronic AMPK stimulation both physiologically and pharmacologically for DM1 patients.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ali Al-Rewashdy
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Guy Bélanger
- Department of Cellular and Molecular Medicine, 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
| |
Collapse
|
58
|
Morriss GR, Rajapakshe K, Huang S, Coarfa C, Cooper TA. Mechanisms of skeletal muscle wasting in a mouse model for myotonic dystrophy type 1. Hum Mol Genet 2019; 27:2789-2804. [PMID: 29771332 DOI: 10.1093/hmg/ddy192] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/14/2018] [Indexed: 12/18/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multi-systemic disease resulting in severe muscle weakening and wasting. DM1 is caused by expansion of CTG repeats in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. We have developed an inducible, skeletal muscle-specific mouse model of DM1 (CUG960) that expresses 960 CUG repeat-expressing animals (CUG960) in the context of human DMPK exons 11-15. CUG960 RNA-expressing mice induced at postnatal day 1, as well as adult-onset animals, show clear, measurable muscle wasting accompanied by severe histological defects including central myonuclei, reduced fiber cross-sectional area, increased percentage of oxidative myofibers, the presence of nuclear RNA foci that colocalize with Mbnl1 protein, and increased Celf1 protein in severely affected muscles. Importantly, muscle loss, histological abnormalities and RNA foci are reversible, demonstrating recovery upon removal of toxic RNA. RNA-seq and protein array analysis indicate that the balance between anabolic and catabolic pathways that normally regulate muscle mass may be disrupted by deregulation of platelet derived growth factor receptor β signaling and the PI3K/AKT pathways, along with prolonged activation of AMP-activated protein kinase α signaling. Similar changes were detected in DM1 skeletal muscle compared with unaffected controls. The mouse model presented in this paper shows progressive skeletal muscle wasting and has been used to identify potential molecular mechanisms underlying skeletal muscle loss. The reversibility of the phenotype establishes a baseline response for testing therapeutic approaches.
Collapse
Affiliation(s)
- Ginny R Morriss
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas A Cooper
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
59
|
Roussel MP, Morin M, Gagnon C, Duchesne E. What is known about the effects of exercise or training to reduce skeletal muscle impairments of patients with myotonic dystrophy type 1? A scoping review. BMC Musculoskelet Disord 2019; 20:101. [PMID: 30836978 PMCID: PMC6402179 DOI: 10.1186/s12891-019-2458-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/06/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is a neuromuscular disease characterized by multisystemic involvements including a progressive loss of maximal muscle strength and muscle wasting. Poor lower-limb strength is an important factor explaining disrupted social participation of affected individuals. This review aims to map what is known about the effects of exercise and training programs undertaken to counteract skeletal muscle impairments in DM1 patients. METHODS Medline, CINAHL and EMBASE databases were searched. Regarding study eligibility, title and abstract of 704 studies followed by 45 full articles were reviewed according to the following eligibility criteria. Inclusion: (1) humans with DM1 and (2) experimental protocol relying on exercise or training. Exclusion: (1) studies that do not evaluate skeletal muscle responses or adaptations, (2) reviews covering articles already included and (3) pharmacological intervention at the same time of exercise or training program. RESULTS Twenty-one papers were selected for in-depth analysis. Different exercise or training protocols were found including: acute exercise, neuromuscular electric stimulation, strength training, aerobic training, balance training and multiple rehabilitation interventions. Seven studies reported clinical measurements only, five physiological parameters only and nine both types. CONCLUSION This scoping review offers a complete summary of the current scientific literature on the effect of exercise and training in DM1 and a framework for future studies based on the concomitant evaluation of the several outcomes in present literature. Although there were a good number of studies focusing on clinical measurements, heterogeneity between studies does not allow to identify what are the adequate training parameters to obtain exercise or training-induced positive impacts on muscle function. Scientific literature is even more scarce regarding physiological parameters, where much more research is needed to understand the underlying mechanisms of exercise response in DM1.
Collapse
Affiliation(s)
- Marie-Pier Roussel
- Département des sciences de la santé, physiothérapie, Université du Québec à Chicoutimi, 555, boulevard de l’Université, Chicoutimi, Quebec G7H 2B1 Canada
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay–Lac-St-Jean, 2230 rue de l’Hôpital, Saguenay, Québec Canada
- Centre de recherche Charles-Le Moyne – Saguenay–Lac-Saint-Jean sur les innovations en santé, 2230 rue de l’Hôpital, Saguenay, Québec, Canada., Longueuil, Québec Canada
| | - Marika Morin
- Département des sciences de la santé, physiothérapie, Université du Québec à Chicoutimi, 555, boulevard de l’Université, Chicoutimi, Quebec G7H 2B1 Canada
| | - Cynthia Gagnon
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay–Lac-St-Jean, 2230 rue de l’Hôpital, Saguenay, Québec Canada
- Centre de recherche Charles-Le Moyne – Saguenay–Lac-Saint-Jean sur les innovations en santé, 2230 rue de l’Hôpital, Saguenay, Québec, Canada., Longueuil, Québec Canada
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12e Avenue Nord, Sherbrooke, Québec Canada
| | - Elise Duchesne
- Département des sciences de la santé, physiothérapie, Université du Québec à Chicoutimi, 555, boulevard de l’Université, Chicoutimi, Quebec G7H 2B1 Canada
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires, Centre intégré universitaire de santé et de services sociaux du Saguenay–Lac-St-Jean, 2230 rue de l’Hôpital, Saguenay, Québec Canada
- Centre de recherche Charles-Le Moyne – Saguenay–Lac-Saint-Jean sur les innovations en santé, 2230 rue de l’Hôpital, Saguenay, Québec, Canada., Longueuil, Québec Canada
| |
Collapse
|
60
|
Manta A, Stouth DW, Xhuti D, Chi L, Rebalka IA, Kalmar JM, Hawke TJ, Ljubicic V. Chronic exercise mitigates disease mechanisms and improves muscle function in myotonic dystrophy type 1 mice. J Physiol 2019; 597:1361-1381. [PMID: 30628727 DOI: 10.1113/jp277123] [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] [Received: 09/09/2018] [Accepted: 01/04/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Myotonic dystrophy type 1 (DM1), the second most common muscular dystrophy and most prevalent adult form of muscular dystrophy, is characterized by muscle weakness, wasting and myotonia. A microsatellite repeat expansion mutation results in RNA toxicity and dysregulation of mRNA processing, which are the primary downstream causes of the disorder. Recent studies with DM1 participants demonstrate that exercise is safe, enjoyable and elicits benefits in muscle strength and function; however, the molecular mechanisms of exercise adaptation in DM1 are undefined. Our results demonstrate that 7 weeks of volitional running wheel exercise in a pre-clinical DM1 mouse model resulted in significantly improved motor performance, muscle strength and endurance, as well as reduced myotonia. At the cellular level, chronic physical activity attenuated RNA toxicity, liberated Muscleblind-like 1 protein from myonuclear foci and improved mRNA alternative splicing. ABSTRACT Myotonic dystrophy type 1 (DM1) is a trinucleotide repeat expansion neuromuscular disorder that is most prominently characterized by skeletal muscle weakness, wasting and myotonia. Chronic physical activity is safe and satisfying, and can elicit functional benefits such as improved strength and endurance in DM1 patients, but the underlying cellular basis of exercise adaptation is undefined. Our purpose was to examine the mechanisms of exercise biology in DM1. Healthy, sedentary wild-type (SED-WT) mice, as well as sedentary human skeletal actin-long repeat animals, a murine model of DM1 myopathy (SED-DM1), and DM1 mice with volitional access to a running wheel for 7 weeks (EX-DM1), were utilized. Chronic exercise augmented strength and endurance in vivo and in situ in DM1 mice. These alterations coincided with normalized measures of myopathy, as well as increased mitochondrial content. Electromyography revealed a 70-85% decrease in the duration of myotonic discharges in muscles from EX-DM1 compared to SED-DM1 animals. The exercise-induced enhancements in muscle function corresponded at the molecular level with mitigated spliceopathy, specifically the processing of bridging integrator 1 and muscle-specific chloride channel (CLC-1) transcripts. CLC-1 protein content and sarcolemmal expression were lower in SED-DM1 versus SED-WT animals, but they were similar between SED-WT and EX-DM1 groups. Chronic exercise also attenuated RNA toxicity, as indicated by reduced (CUG)n foci-positive myonuclei and sequestered Muscleblind-like 1 (MBNL1). Our data indicate that chronic exercise-induced physiological improvements in DM1 occur in concert with mitigated primary downstream disease mechanisms, including RNA toxicity, MBNL1 loss-of-function, and alternative mRNA splicing.
Collapse
Affiliation(s)
- Alexander Manta
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Derek W Stouth
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Donald Xhuti
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Leon Chi
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Jayne M Kalmar
- Department of Kinesiology & Physical Education, Wilfred Laurier University, Waterloo, ON, Canada, N2L 3C5
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| |
Collapse
|
61
|
Maury Y, Poydenot P, Brinon B, Lesueur L, Gide J, Roquevière S, Côme J, Polvèche H, Auboeuf D, Alexandre Denis J, Pietu G, Furling D, Lechuga M, Baghdoyan S, Peschanski M, Martinat C. Pluripotent Stem Cell-Based Drug Screening Reveals Cardiac Glycosides as Modulators of Myotonic Dystrophy Type 1. iScience 2019; 11:258-271. [PMID: 30639849 PMCID: PMC6327858 DOI: 10.1016/j.isci.2018.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
There is currently no treatment for myotonic dystrophy type 1 (DM1), the most frequent myopathy of genetic origin. This progressive neuromuscular disease is caused by nuclear-retained RNAs containing expanded CUG repeats. These toxic RNAs alter the activities of RNA splicing factors, resulting in alternative splicing misregulation. By combining human mutated pluripotent stem cells and phenotypic drug screening, we revealed that cardiac glycosides act as modulators for both upstream nuclear aggregations of DMPK mRNAs and several downstream alternative mRNA splicing defects. However, these occurred at different drug concentration ranges. Similar biological effects were recorded in a DM1 mouse model. At the mechanistic level, we demonstrated that this effect was calcium dependent and was synergic with inhibition of the ERK pathway. These results further underscore the value of stem-cell-based assays for drug discovery in monogenic diseases. Myotonic dystrophy type 1 hPSCs were adapted for high content screening FDA-approved cardiac glycosides normalize in vitro and in vivo DM1 biological markers Cardiac glycosides synergize with the ERK pathway to normalize DM1 biomarkers This study emphasizes the value of human pluripotent stem cells for drug discovery
Collapse
Affiliation(s)
- Yves Maury
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | - Lea Lesueur
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | - Julien Côme
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | | | - Geneviève Pietu
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | - Denis Furling
- Sorbonne Universités UPMC Univ Paris 06, INSERM, Centre de Recherche en Myologie - UMRS974, Institut de Myologie, 75013 Paris, France
| | - Marc Lechuga
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | - Marc Peschanski
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France; INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | - Cécile Martinat
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France.
| |
Collapse
|
62
|
Abstract
Myotonic dystrophy is an autosomal dominant muscular dystrophy not only associated with muscle weakness, atrophy, and myotonia but also prominent multisystem involvement. There are 2 similar, but distinct, forms of myotonic dystrophy; type 1 is caused by a CTG repeat expansion in the DMPK gene, and type 2 is caused by a CCTG repeat expansion in the CNBP gene. Type 1 is associated with distal limb, neck flexor, and bulbar weakness and results in different phenotypic subtypes with variable onset from congenital to very late-onset as well as variable signs and symptoms. The classically described adult-onset form is the most common. In contrast, myotonic dystrophy type 2 is adult-onset or late-onset, has proximal predominant muscle weakness, and generally has less severe multisystem involvement. In both forms of myotonic dystrophy, the best characterized disease mechanism is a RNA toxic gain-of-function during which RNA repeats form nuclear foci resulting in sequestration of RNA-binding proteins and, therefore, dysregulated splicing of premessenger RNA. There are currently no disease-modifying therapies, but clinical surveillance, preventative measures, and supportive treatments are used to reduce the impact of muscular impairment and other systemic involvement including cataracts, cardiac conduction abnormalities, fatigue, central nervous system dysfunction, respiratory weakness, dysphagia, and endocrine dysfunction. Exciting preclinical progress has been made in identifying a number of potential strategies including genome editing, small molecule therapeutics, and antisense oligonucleotide-based therapies to target the pathogenesis of type 1 and type 2 myotonic dystrophies at the DNA, RNA, or downstream target level.
Collapse
Affiliation(s)
- Samantha LoRusso
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, Columbus, OH, 43210, USA
| | - Benjamin Weiner
- The Ohio State University College of Medicine, The Ohio State University, 370 West 9th Avenue, Columbus, OH, 43210, USA
| | - W David Arnold
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
63
|
Zapata-Aldana E, Ceballos-Sáenz D, Hicks R, Campbell C. Prenatal, Neonatal, and Early Childhood Features in Congenital Myotonic Dystrophy. J Neuromuscul Dis 2018; 5:331-340. [DOI: 10.3233/jnd-170277] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Eugenio Zapata-Aldana
- Division of Pediatric Neurology, Children’s Hospital London Health Science Centre London, ON, Canada
| | - Delia Ceballos-Sáenz
- Division of Pediatric Neurology, Children’s Hospital London Health Science Centre London, ON, Canada
| | - Rhiannon Hicks
- Division of Pediatric Neurology, Children’s Hospital London Health Science Centre London, ON, Canada
| | - Craig Campbell
- Division of Pediatric Neurology, Children’s Hospital London Health Science Centre London, ON, Canada
- Clinical Neurological Sciences, Western University, London, ON, Canada
- Epidemiology, Western University, London, ON, Canada
| |
Collapse
|
64
|
Braz SO, Acquaire J, Gourdon G, Gomes-Pereira M. Of Mice and Men: Advances in the Understanding of Neuromuscular Aspects of Myotonic Dystrophy. Front Neurol 2018; 9:519. [PMID: 30050493 PMCID: PMC6050950 DOI: 10.3389/fneur.2018.00519] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 06/12/2018] [Indexed: 12/26/2022] Open
Abstract
Intensive effort has been directed toward the modeling of myotonic dystrophy (DM) in mice, in order to reproduce human disease and to provide useful tools to investigate molecular and cellular pathogenesis and test efficient therapies. Mouse models have contributed to dissect the multifaceted impact of the DM mutation in various tissues, cell types and in a pleiotropy of pathways, through the expression of toxic RNA transcripts. Changes in alternative splicing, transcription, translation, intracellular RNA localization, polyadenylation, miRNA metabolism and phosphorylation of disease intermediates have been described in different tissues. Some of these events have been directly associated with specific disease symptoms in the skeletal muscle and heart of mice, offering the molecular explanation for individual disease phenotypes. In the central nervous system (CNS), however, the situation is more complex. We still do not know how the molecular abnormalities described translate into CNS dysfunction, nor do we know if the correction of individual molecular events will provide significant therapeutic benefits. The variability in model design and phenotypes described so far requires a thorough and critical analysis. In this review we discuss the recent contributions of mouse models to the understanding of neuromuscular aspects of disease, therapy development, and we provide a reflective assessment of our current limitations and pressing questions that remain unanswered.
Collapse
Affiliation(s)
- Sandra O Braz
- Laboratory CTGDM, INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Julien Acquaire
- Laboratory CTGDM, INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Geneviève Gourdon
- Laboratory CTGDM, INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Mário Gomes-Pereira
- Laboratory CTGDM, INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| |
Collapse
|
65
|
Reduction of Cellular Nucleic Acid Binding Protein Encoded by a Myotonic Dystrophy Type 2 Gene Causes Muscle Atrophy. Mol Cell Biol 2018; 38:MCB.00649-17. [PMID: 29735719 DOI: 10.1128/mcb.00649-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/26/2018] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 2 (DM2) is a neuromuscular disease caused by an expansion of intronic CCTG repeats in the CNBP gene, which encodes a protein regulating translation and transcription. To better understand the role of cellular nucleic acid binding protein (CNBP) in DM2 pathology, we examined skeletal muscle in a new model of Cnbp knockout (KO) mice. This study showed that a loss of Cnbp disturbs myofibrillar sarcomeric organization at birth. Surviving homozygous Cnbp KO mice develop muscle atrophy at a young age. The skeletal muscle phenotype in heterozygous Cnbp KO mice was milder, but they developed severe muscle wasting at an advanced age. Several proteins that control global translation and muscle contraction are altered in muscle of Cnbp KO mice. A search for CNBP binding proteins showed that CNBP interacts with the α subunit of the dystroglycan complex, a core component of the multimeric dystrophin-glycoprotein complex, which regulates membrane stability. Whereas CNBP is reduced in cytoplasm of DM2 human fibers, it is a predominantly membrane protein in DM2 fibers, and its interaction with α-dystroglycan is increased in DM2. These findings suggest that alterations of CNBP in DM2 might cause muscle atrophy via CNBP-mediated translation and via protein-protein interactions affecting myofiber membrane function.
Collapse
|
66
|
Cerro-Herreros E, Sabater-Arcis M, Fernandez-Costa JM, Moreno N, Perez-Alonso M, Llamusi B, Artero R. miR-23b and miR-218 silencing increase Muscleblind-like expression and alleviate myotonic dystrophy phenotypes in mammalian models. Nat Commun 2018; 9:2482. [PMID: 29946070 PMCID: PMC6018771 DOI: 10.1038/s41467-018-04892-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 05/30/2018] [Indexed: 12/16/2022] Open
Abstract
Functional depletion of the alternative splicing factors Muscleblind-like (MBNL 1 and 2) is at the basis of the neuromuscular disease myotonic dystrophy type 1 (DM1). We previously showed the efficacy of miRNA downregulation in Drosophila DM1 model. Here, we screen for miRNAs that regulate MBNL1 and MBNL2 in HeLa cells. We thus identify miR-23b and miR-218, and confirm that they downregulate MBNL proteins in this cell line. Antagonists of miR-23b and miR-218 miRNAs enhance MBNL protein levels and rescue pathogenic missplicing events in DM1 myoblasts. Systemic delivery of these "antagomiRs" similarly boost MBNL expression and improve DM1-like phenotypes, including splicing alterations, histopathology, and myotonia in the HSALR DM1 model mice. These mammalian data provide evidence for therapeutic blocking of the miRNAs that control Muscleblind-like protein expression in myotonic dystrophy.
Collapse
Affiliation(s)
- Estefania Cerro-Herreros
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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
| | - Maria Sabater-Arcis
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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 Fernandez-Costa
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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
| | - Nerea Moreno
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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
| | - Manuel Perez-Alonso
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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
| | - Beatriz Llamusi
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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.
| | - Ruben Artero
- Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI 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.
| |
Collapse
|
67
|
López-Morató M, Brook JD, Wojciechowska M. Small Molecules Which Improve Pathogenesis of Myotonic Dystrophy Type 1. Front Neurol 2018; 9:349. [PMID: 29867749 PMCID: PMC5968088 DOI: 10.3389/fneur.2018.00349] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/30/2018] [Indexed: 12/30/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults for which there is currently no treatment. The pathogenesis of this autosomal dominant disorder is associated with the expansion of CTG repeats in the 3'-UTR of the DMPK gene. DMPK transcripts with expanded CUG repeats (CUGexpDMPK) are retained in the nucleus forming multiple discrete foci, and their presence triggers a cascade of toxic events. Thus far, most research emphasis has been on interactions of CUGexpDMPK with the muscleblind-like (MBNL) family of splicing factors. These proteins are sequestered by the expanded CUG repeats of DMPK RNA leading to their functional depletion. As a consequence, abnormalities in many pathways of RNA metabolism, including alternative splicing, are detected in DM1. To date, in vitro and in vivo efforts to develop therapeutic strategies for DM1 have mostly been focused on targeting CUGexpDMPK via reducing their expression and/or preventing interactions with MBNL1. Antisense oligonucleotides targeted to the CUG repeats in the DMPK transcripts are of particular interest due to their potential capacity to discriminate between mutant and normal transcripts. However, a growing number of reports describe alternative strategies using small molecule chemicals acting independently of a direct interaction with CUGexpDMPK. In this review, we summarize current knowledge about these chemicals and we describe the beneficial effects they caused in different DM1 experimental models. We also present potential mechanisms of action of these compounds and pathways they affect which could be considered for future therapeutic interventions in DM1.
Collapse
Affiliation(s)
- Marta López-Morató
- Queen’s Medical Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - John David Brook
- Queen’s Medical Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Marzena Wojciechowska
- Queen’s Medical Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
- Polish Academy of Sciences, Department of Molecular Genetics, Institute of Bioorganic Chemistry, Poznan, Poland
| |
Collapse
|
68
|
Witherick J, Brady S. Update on muscle disease. J Neurol 2018; 265:1717-1725. [DOI: 10.1007/s00415-018-8856-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 03/30/2018] [Indexed: 12/24/2022]
|
69
|
Thomas JD, Oliveira R, Sznajder ŁJ, Swanson MS. Myotonic Dystrophy and Developmental Regulation of RNA Processing. Compr Physiol 2018; 8:509-553. [PMID: 29687899 PMCID: PMC11323716 DOI: 10.1002/cphy.c170002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myotonic dystrophy (DM) is a multisystemic disorder caused by microsatellite expansion mutations in two unrelated genes leading to similar, yet distinct, diseases. DM disease presentation is highly variable and distinguished by differences in age-of-onset and symptom severity. In the most severe form, DM presents with congenital onset and profound developmental defects. At the molecular level, DM pathogenesis is characterized by a toxic RNA gain-of-function mechanism that involves the transcription of noncoding microsatellite expansions. These mutant RNAs disrupt key cellular pathways, including RNA processing, localization, and translation. In DM, these toxic RNA effects are predominantly mediated through the modulation of the muscleblind-like and CUGBP and ETR-3-like factor families of RNA binding proteins (RBPs). Dysfunction of these RBPs results in widespread RNA processing defects culminating in the expression of developmentally inappropriate protein isoforms in adult tissues. The tissue that is the focus of this review, skeletal muscle, is particularly sensitive to mutant RNA-responsive perturbations, as patients display a variety of developmental, structural, and functional defects in muscle. Here, we provide a comprehensive overview of DM1 and DM2 clinical presentation and pathology as well as the underlying cellular and molecular defects associated with DM disease onset and progression. Additionally, fundamental aspects of skeletal muscle development altered in DM are highlighted together with ongoing and potential therapeutic avenues to treat this muscular dystrophy. © 2018 American Physiological Society. Compr Physiol 8:509-553, 2018.
Collapse
Affiliation(s)
- James D. Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Ruan Oliveira
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Łukasz J. Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Maurice S. Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
70
|
Wei C, Stock L, Valanejad L, Zalewski ZA, Karns R, Puymirat J, Nelson D, Witte D, Woodgett J, Timchenko NA, Timchenko L. Correction of GSK3β at young age prevents muscle pathology in mice with myotonic dystrophy type 1. FASEB J 2018; 32:2073-2085. [PMID: 29203592 DOI: 10.1096/fj.201700700r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is a progressive neuromuscular disease caused by expanded CUG repeats, which misregulate RNA metabolism through several RNA-binding proteins, including CUG-binding protein/CUGBP1 elav-like factor 1 (CUGBP1/CELF1) and muscleblind 1 protein. Mutant CUG repeats elevate CUGBP1 and alter CUGBP1 activity via a glycogen synthase kinase 3β (GSK3β)-cyclin D3-cyclin D-dependent kinase 4 (CDK4) signaling pathway. Inhibition of GSK3β corrects abnormal activity of CUGBP1 in DM1 mice [human skeletal actin mRNA, containing long repeats ( HSALR) model]. Here, we show that the inhibition of GSK3β in young HSALR mice prevents development of DM1 muscle pathology. Skeletal muscle in 1-yr-old HSALR mice, treated at 1.5 mo for 6 wk with the inhibitors of GSK3, exhibits high fiber density, corrected atrophy, normal fiber size, with reduced central nuclei and normalized grip strength. Because CUG-GSK3β-cyclin D3-CDK4 converts the active form of CUGBP1 into a form of translational repressor, we examined the contribution of CUGBP1 in myogenesis using Celf1 knockout mice. We found that a loss of CUGBP1 disrupts myogenesis, affecting genes that regulate differentiation and the extracellular matrix. Proteins of those pathways are also misregulated in young HSALR mice and in muscle biopsies of patients with congenital DM1. These findings suggest that the correction of GSK3β-CUGBP1 pathway in young HSALR mice might have a positive effect on the myogenesis over time.-Wei, C., Stock, L., Valanejad, L., Zalewski, Z. A., Karns, R., Puymirat, J., Nelson, D., Witte, D., Woodgett, J., Timchenko, N. A., Timchenko, L. Correction of GSK3β at young age prevents muscle pathology in mice with myotonic dystrophy type 1.
Collapse
Affiliation(s)
- Christina Wei
- Division of Neurology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Lauren Stock
- Division of Neurology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Leila Valanejad
- Department of Surgery, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Zachary A Zalewski
- Department of Molecular Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Rebekah Karns
- Department of Bioinformatics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Jack Puymirat
- Centre Hospitalier-Université Laval Research Center, Québec City, Quebéc, Canada
| | - David Nelson
- Department of Molecular Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - David Witte
- Department of Pathology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA; and
| | - Jim Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Nikolai A Timchenko
- Department of Surgery, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Lubov Timchenko
- Division of Neurology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| |
Collapse
|
71
|
Ravel-Chapuis A, Bélanger G, Côté J, Michel RN, Jasmin BJ. Misregulation of calcium-handling proteins promotes hyperactivation of calcineurin-NFAT signaling in skeletal muscle of DM1 mice. Hum Mol Genet 2017; 26:2192-2206. [PMID: 28369518 DOI: 10.1093/hmg/ddx109] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/16/2017] [Indexed: 12/26/2022] Open
Abstract
Myotonic Dystrophy type 1 (DM1) is caused by an expansion of CUG repeats in DMPK mRNAs. This mutation affects alternative splicing through misregulation of RNA-binding proteins. Amongst pre-mRNAs that are mis-spliced, several code for proteins involved in calcium homeostasis suggesting that calcium-handling and signaling are perturbed in DM1. Here, we analyzed expression of such proteins in DM1 mouse muscle. We found that the levels of several sarcoplasmic reticulum proteins (SERCA1, sarcolipin and calsequestrin) are altered, likely contributing to an imbalance in calcium homeostasis. We also observed that calcineurin (CnA) signaling is hyperactivated in DM1 muscle. Indeed, CnA expression and phosphatase activity are both markedly increased in DM1 muscle. Coherent with this, we found that activators of the CnA pathway (MLP, FHL1) are also elevated. Consequently, NFATc1 expression is increased in DM1 muscle and becomes relocalized to myonuclei, together with an up-regulation of its transcriptional targets (RCAN1.4 and myoglobin). Accordingly, DM1 mouse muscles display an increase in oxidative metabolism and fiber hypertrophy. To determine the functional consequences of this CnA hyperactivation, we administered cyclosporine A, an inhibitor of CnA, to DM1 mice. Muscles of treated DM1 mice showed an increase in CUGBP1 levels, and an exacerbation of key alternative splicing events associated with DM1. Finally, inhibition of CnA in cultured human DM1 myoblasts also resulted in a splicing exacerbation of the insulin receptor. Together, these findings show for the first time that calcium-CnA signaling is hyperactivated in DM1 muscle and that such hyperactivation represents a beneficial compensatory adaptation to the disease.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine and Center for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Guy Bélanger
- Department of Cellular and Molecular Medicine and Center for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jocelyn Côté
- Department of Cellular and Molecular Medicine and Center for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Robin N Michel
- Department of Exercise Science, Faculty of Arts and Science, Concordia University, Montreal, QC, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine and Center for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
72
|
Renna LV, Bosè F, Iachettini S, Fossati B, Saraceno L, Milani V, Colombo R, Meola G, Cardani R. Receptor and post-receptor abnormalities contribute to insulin resistance in myotonic dystrophy type 1 and type 2 skeletal muscle. PLoS One 2017; 12:e0184987. [PMID: 28915272 PMCID: PMC5600405 DOI: 10.1371/journal.pone.0184987] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/04/2017] [Indexed: 12/27/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant multisystemic disorders caused by expansion of microsatellite repeats. In both forms, the mutant transcripts accumulate in nuclear foci altering the function of alternative splicing regulators which are necessary for the physiological mRNA processing. Missplicing of insulin receptor (IR) gene (INSR) has been associated with insulin resistance, however, it cannot be excluded that post-receptor signalling abnormalities could also contribute to this feature in DM. We have analysed the insulin pathway in skeletal muscle biopsies and in myotube cultures from DM patients to assess whether downstream metabolism might be dysregulated and to better characterize the mechanism inducing insulin resistance. DM skeletal muscle exhibits alterations of basal phosphorylation levels of Akt/PKB, p70S6K, GSK3β and ERK1/2, suggesting that these changes might be accompanied by a lack of further insulin stimulation. Alterations of insulin pathway have been confirmed on control and DM myotubes expressing fetal INSR isoform (INSR-A). The results indicate that insulin action appears to be lower in DM than in control myotubes in terms of protein activation and glucose uptake. Our data indicate that post-receptor signalling abnormalities might contribute to DM insulin resistance regardless the alteration of INSR splicing.
Collapse
Affiliation(s)
- Laura Valentina Renna
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Francesca Bosè
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Sara Iachettini
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Barbara Fossati
- Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Lorenzo Saraceno
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Valentina Milani
- Scientific Directorate, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Roberto Colombo
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giovanni Meola
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy.,Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| |
Collapse
|
73
|
Konieczny P, Selma-Soriano E, Rapisarda AS, Fernandez-Costa JM, Perez-Alonso M, Artero R. Myotonic dystrophy: candidate small molecule therapeutics. Drug Discov Today 2017; 22:1740-1748. [PMID: 28780071 DOI: 10.1016/j.drudis.2017.07.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/14/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is a rare multisystemic neuromuscular disorder caused by expansion of CTG trinucleotide repeats in the noncoding region of the DMPK gene. Mutant DMPK transcripts are toxic and alter gene expression at several levels. Chiefly, the secondary structure formed by CUGs has a strong propensity to capture and retain proteins, like those of the muscleblind-like (MBNL) family. Sequestered MBNL proteins cannot then fulfill their normal functions. Many therapeutic approaches have been explored to reverse these pathological consequences. Here, we review the myriad of small molecules that have been proposed for DM1, including examples obtained from computational rational design, HTS, drug repurposing, and therapeutic gene modulation.
Collapse
Affiliation(s)
- Piotr Konieczny
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| | - Estela Selma-Soriano
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| | - Anna S Rapisarda
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| | - Juan M Fernandez-Costa
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| | - Manuel Perez-Alonso
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain
| | - Ruben Artero
- Department of Genetics and Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), Universitat de València, Valencia, Spain; Translational Genomics Group, Incliva Health Research Institute, Valencia, Spain; Joint Unit Incliva-CIPF, Valencia, Spain.
| |
Collapse
|
74
|
Palomo V, Perez DI, Roca C, Anderson C, Rodríguez-Muela N, Perez C, Morales-Garcia JA, Reyes JA, Campillo NE, Perez-Castillo AM, Rubin LL, Timchenko L, Gil C, Martinez A. Subtly Modulating Glycogen Synthase Kinase 3 β: Allosteric Inhibitor Development and Their Potential for the Treatment of Chronic Diseases. J Med Chem 2017; 60:4983-5001. [PMID: 28548834 DOI: 10.1021/acs.jmedchem.7b00395] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glycogen synthase kinase 3 β (GSK-3β) is a central target in several unmet diseases. To increase the specificity of GSK-3β inhibitors in chronic treatments, we developed small molecules allowing subtle modulation of GSK-3β activity. Design synthesis, structure-activity relationships, and binding mode of quinoline-3-carbohydrazide derivatives as allosteric modulators of GSK-3β are presented here. Furthermore, we show how allosteric binders may overcome the β-catenin side effects associated with strong GSK-3β inhibition. The therapeutic potential of some of these modulators has been tested in human samples from patients with congenital myotonic dystrophy type 1 (CDM1) and spinal muscular atrophy (SMA) patients. We found that compound 53 improves delayed myogenesis in CDM1 myoblasts, while compounds 1 and 53 have neuroprotective properties in SMA-derived cells. These findings suggest that the allosteric modulators of GSK-3β may be used for future development of drugs for DM1, SMA, and other chronic diseases where GSK-3β inhibition exhibits therapeutic effects.
Collapse
Affiliation(s)
- Valle Palomo
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Daniel I Perez
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Carlos Roca
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Cara Anderson
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Hospital , Cincinnati, Ohio 45219, United States
| | - Natalia Rodríguez-Muela
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts 02138, United States
| | - Concepción Perez
- Instituto de Quimica Médica-CSIC , Juan del Cierva 3, 28006 Madrid, Spain
| | - Jose A Morales-Garcia
- Instituto de Investigaciones Biomedicas-CSIC , Arturo Duperier 4, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Calle de Valderrebollo 5, 28031 Madrid, Spain
| | - Julio A Reyes
- Instituto de Quimica Médica-CSIC , Juan del Cierva 3, 28006 Madrid, Spain
| | - Nuria E Campillo
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ana M Perez-Castillo
- Instituto de Investigaciones Biomedicas-CSIC , Arturo Duperier 4, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Calle de Valderrebollo 5, 28031 Madrid, Spain
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology and Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts 02138, United States
| | - Lubov Timchenko
- Department of Pediatrics, Division of Neurology, Cincinnati Children's Hospital , Cincinnati, Ohio 45219, United States
| | - Carmen Gil
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas-CSIC , Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
75
|
Thornton CA, Wang E, Carrell EM. Myotonic dystrophy: approach to therapy. Curr Opin Genet Dev 2017; 44:135-140. [PMID: 28376341 PMCID: PMC5447481 DOI: 10.1016/j.gde.2017.03.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/25/2017] [Accepted: 03/13/2017] [Indexed: 01/16/2023]
Abstract
Myotonic dystrophy (DM) is a dominantly-inherited genetic disorder affecting skeletal muscle, heart, brain, and other organs. DM type 1 is caused by expansion of a CTG triplet repeat in DMPK, whereas DM type 2 is caused by expansion of a CCTG tetramer repeat in CNBP. In both cases the DM mutations lead to expression of dominant-acting RNAs. Studies of RNA toxicity have now revealed novel mechanisms and new therapeutic targets. Preclinical data have suggested that RNA dominance is responsive to therapeutic intervention and that DM therapy can be approached at several different levels. Here we review recent efforts to alleviate RNA toxicity in DM.
Collapse
Affiliation(s)
- Charles A Thornton
- Department of Neurology, University of Rochester, Rochester 14642, NY, United States.
| | - Eric Wang
- Department of Molecular Genetics & Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, FL, United States
| | - Ellie M Carrell
- Department of Neurology, University of Rochester, Rochester 14642, NY, United States
| |
Collapse
|
76
|
Gourdon G, Meola G. Myotonic Dystrophies: State of the Art of New Therapeutic Developments for the CNS. Front Cell Neurosci 2017; 11:101. [PMID: 28473756 PMCID: PMC5397409 DOI: 10.3389/fncel.2017.00101] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022] Open
Abstract
Myotonic dystrophies are multisystemic diseases characterized not only by muscle and heart dysfunction but also by CNS alteration. They are now recognized as brain diseases affecting newborns and children for myotonic dystrophy type 1 and adults for both myotonic dystrophy type 1 and type 2. In the past two decades, much progress has been made in understanding the mechanisms underlying the DM symptoms allowing development of new molecular therapeutic tools with the ultimate aim of curing the disease. This review describes the state of the art for the characterization of CNS related symptoms, the development of molecular strategies to target the CNS as well as the available tools for screening and testing new possible treatments.
Collapse
Affiliation(s)
- Genevieve Gourdon
- Institut National de la Santé et de la Recherche Médicale UMR1163Paris, France.,Laboratory CTGDM, Institut Imagine, Université Paris Descartes-Sorbonne Paris CitéParis, France
| | - Giovanni Meola
- Department of Biomedical Sciences for Health, Policlinico San Donato (IRCCS), University of MilanMilan, Italy
| |
Collapse
|
77
|
Shen C, Zhou J, Wang X, Yu XY, Liang C, Liu B, Pan X, Zhao Q, Song JL, Wang J, Bao M, Wu C, Li Y, Song YH. Angiotensin-II-induced Muscle Wasting is Mediated by 25-Hydroxycholesterol via GSK3β Signaling Pathway. EBioMedicine 2017; 16:238-250. [PMID: 28161398 PMCID: PMC5474518 DOI: 10.1016/j.ebiom.2017.01.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 01/20/2017] [Accepted: 01/27/2017] [Indexed: 12/14/2022] Open
Abstract
While angiotensin II (ang II) has been implicated in the pathogenesis of cardiac cachexia (CC), the molecules that mediate ang II's wasting effect have not been identified. It is known TNF-α level is increased in patients with CC, and TNF-α release is triggered by ang II. We therefore hypothesized that ang II induced muscle wasting is mediated by TNF-α. Ang II infusion led to skeletal muscle wasting in wild type (WT) but not in TNF alpha type 1 receptor knockout (TNFR1KO) mice, suggesting that ang II induced muscle loss is mediated by TNF-α through its type 1 receptor. Microarray analysis identified cholesterol 25-hydroxylase (Ch25h) as the down stream target of TNF-α. Intraperitoneal injection of 25-hydroxycholesterol (25-OHC), the product of Ch25h, resulted in muscle loss in C57BL/6 mice, accompanied by increased expression of atrogin-1, MuRF1 and suppression of IGF-1/Akt signaling pathway. The identification of 25-OHC as an inducer of muscle wasting has implications for the development of specific treatment strategies in preventing muscle loss. Ang II induced muscle wasting is mediated by TNF-α, which in turn up regulates Ch25h Knockout of TNFR1 inhibits the production of 25-OHC and blocks ang II induced muscle loss in mice 25-OHC injection induces muscle wasting in mice by activating GSK3β A GSK3β inhibitor blocks ang II induced muscle atrophy, which paves the way for targeted therapy to treat muscle wasting
Cardiac cachexia (CC), a condition characterized by weight loss and muscle wasting, is a serious complication that occurs in patients with chronic heart failure. This condition impairs patient's daily physical activity and their quality of life. Specific therapy for CC is currently unavailable because the pathogenesis remains unknown. Previous studies have identified angiotensin II (ang II) as an important mediator of CC. We now report a previously unrecognized role of 25-hydroxycholesterol (25-OHC) in mediating ang II induced muscle loss. The identification of 25-OHC as a muscle wasting inducer has implications for the development of therapeutic intervention in preserving muscle mass.
Collapse
Affiliation(s)
- Congcong Shen
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, PR China.
| | - Xiaoxiao Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Xi-Yong Yu
- Guangdong Cardiovascular Institute, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chun Liang
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, No. 415, Fengyang Road, Shanghai, PR China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Ji Lin University, Changchun, Jilin 130021, PR China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, PR China
| | - Qiong Zhao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jenny Lee Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Jiajun Wang
- Department of Gynecology, The Affiliated Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, PR China
| | - Meiyu Bao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Chaofan Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China
| | - Yangxin Li
- The Department of Cardiovascular Surgery of the First Affiliated Hospital and the Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Yao-Hua Song
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, PR China.
| |
Collapse
|
78
|
Brockhoff M, Rion N, Chojnowska K, Wiktorowicz T, Eickhorst C, Erne B, Frank S, Angelini C, Furling D, Rüegg MA, Sinnreich M, Castets P. Targeting deregulated AMPK/mTORC1 pathways improves muscle function in myotonic dystrophy type I. J Clin Invest 2017; 127:549-563. [PMID: 28067669 DOI: 10.1172/jci89616] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
Myotonic dystrophy type I (DM1) is a disabling multisystemic disease that predominantly affects skeletal muscle. It is caused by expanded CTG repeats in the 3'-UTR of the dystrophia myotonica protein kinase (DMPK) gene. RNA hairpins formed by elongated DMPK transcripts sequester RNA-binding proteins, leading to mis-splicing of numerous pre-mRNAs. Here, we have investigated whether DM1-associated muscle pathology is related to deregulation of central metabolic pathways, which may identify potential therapeutic targets for the disease. In a well-characterized mouse model for DM1 (HSALR mice), activation of AMPK signaling in muscle was impaired under starved conditions, while mTORC1 signaling remained active. In parallel, autophagic flux was perturbed in HSALR muscle and in cultured human DM1 myotubes. Pharmacological approaches targeting AMPK/mTORC1 signaling greatly ameliorated muscle function in HSALR mice. AICAR, an AMPK activator, led to a strong reduction of myotonia, which was accompanied by partial correction of misregulated alternative splicing. Rapamycin, an mTORC1 inhibitor, improved muscle relaxation and increased muscle force in HSALR mice without affecting splicing. These findings highlight the involvement of AMPK/mTORC1 deregulation in DM1 muscle pathophysiology and may open potential avenues for the treatment of this disease.
Collapse
|
79
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is an unusual protein-serine kinase in that it is primarily regulated by inhibition and lies downstream of multiple cell signaling pathways. This raises a variety of questions in terms of its physiological role(s), how signaling specificity is maintained and why so many eggs have been placed into one basket. There are actually two baskets, as there are two isoforms, GSK-3α and β, that are highly related and largely redundant. Their many substrates range from regulators of cellular metabolism to molecules that control growth and differentiation. In this chapter, we review the characteristics of GSK-3, update progress in understanding the kinase, and try to answer some of the questions raised by its unusual properties. Indeed, the kinase may trigger transformation in our thinking of how cellular signals are organized and controlled.
Collapse
|
80
|
Tajhya RB, Hu X, Tanner MR, Huq R, Kongchan N, Neilson JR, Rodney GG, Horrigan FT, Timchenko LT, Beeton C. Functional KCa1.1 channels are crucial for regulating the proliferation, migration and differentiation of human primary skeletal myoblasts. Cell Death Dis 2016; 7:e2426. [PMID: 27763639 PMCID: PMC5133989 DOI: 10.1038/cddis.2016.324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 01/14/2023]
Abstract
Myoblasts are mononucleated precursors of myofibers; they persist in mature skeletal muscles for growth and regeneration post injury. During myotonic dystrophy type 1 (DM1), a complex autosomal-dominant neuromuscular disease, the differentiation of skeletal myoblasts into functional myotubes is impaired, resulting in muscle wasting and weakness. The mechanisms leading to this altered differentiation are not fully understood. Here, we demonstrate that the calcium- and voltage-dependent potassium channel, KCa1.1 (BK, Slo1, KCNMA1), regulates myoblast proliferation, migration, and fusion. We also show a loss of plasma membrane expression of the pore-forming α subunit of KCa1.1 in DM1 myoblasts. Inhibiting the function of KCa1.1 in healthy myoblasts induced an increase in cytosolic calcium levels and altered nuclear factor kappa B (NFκB) levels without affecting cell survival. In these normal cells, KCa1.1 block resulted in enhanced proliferation and decreased matrix metalloproteinase secretion, migration, and myotube fusion, phenotypes all observed in DM1 myoblasts and associated with disease pathogenesis. In contrast, introducing functional KCa1.1 α-subunits into DM1 myoblasts normalized their proliferation and rescued expression of the late myogenic marker Mef2. Our results identify KCa1.1 channels as crucial regulators of skeletal myogenesis and suggest these channels as novel therapeutic targets in DM1.
Collapse
Affiliation(s)
- Rajeev B Tajhya
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xueyou Hu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark R Tanner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Redwan Huq
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Natee Kongchan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel R Neilson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Frank T Horrigan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lubov T Timchenko
- Department of Pediatrics Neurology, Cincinnati Children's Hospital, Cincinnati, OH 45219, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA.,Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
81
|
Kim YK, Yadava RS, Mandal M, Mahadevan K, Yu Q, Leitges M, Mahadevan MS. Disease Phenotypes in a Mouse Model of RNA Toxicity Are Independent of Protein Kinase Cα and Protein Kinase Cβ. PLoS One 2016; 11:e0163325. [PMID: 27657532 PMCID: PMC5033491 DOI: 10.1371/journal.pone.0163325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023] Open
Abstract
Myotonic dystrophy type 1(DM1) is the prototype for diseases caused by RNA toxicity. RNAs from the mutant allele contain an expanded (CUG)n tract within the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. The toxic RNAs affect the function of RNA binding proteins leading to sequestration of muscleblind-like (MBNL) proteins and increased levels of CELF1 (CUGBP, Elav-like family member 1). The mechanism for increased CELF1 is not very clear. One favored proposition is hyper-phosphorylation of CELF1 by Protein Kinase C alpha (PKCα) leading to increased CELF1 stability. However, most of the evidence supporting a role for PKC-α relies on pharmacological inhibition of PKC. To further investigate the role of PKCs in the pathogenesis of RNA toxicity, we generated transgenic mice with RNA toxicity that lacked both the PKCα and PKCβ isoforms. We find that these mice show similar disease progression as mice wildtype for the PKC isoforms. Additionally, the expression of CELF1 is also not affected by deficiency of PKCα and PKCβ in these RNA toxicity mice. These data suggest that disease phenotypes of these RNA toxicity mice are independent of PKCα and PKCβ.
Collapse
Affiliation(s)
- Yun K. Kim
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ramesh S. Yadava
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mahua Mandal
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Karunasai Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Qing Yu
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael Leitges
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | - Mani S. Mahadevan
- Department of Pathology, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
82
|
Chardon JW, Jasmin BJ, Kothary R, Parks RJ. Report on the 3rd Ottawa International Conference on Neuromuscular Biology, Disease and Therapy - September 24-26, 2015, Ottawa, Canada. J Neuromuscul Dis 2016; 3:431-442. [PMID: 27854234 PMCID: PMC5123627 DOI: 10.3233/jnd-169001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jodi Warman Chardon
- Department of Medicine, The Ottawa Hospital and University of Ottawa.,Centre for Neuromuscular Disease, University of Ottawa.,Department of Pediatrics (Genetics), Children's Hospital of Eastern Ontario.,Neurosciences and Clinical Epidemiology Programs, Ottawa Hospital Research Institute
| | - Bernard J Jasmin
- Centre for Neuromuscular Disease, University of Ottawa.,Department of Cellular and Molecular Medicine, University of Ottawa
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute.,Department of Medicine, The Ottawa Hospital and University of Ottawa.,Centre for Neuromuscular Disease, University of Ottawa.,Department of Cellular and Molecular Medicine, University of Ottawa
| | - Robin J Parks
- Regenerative Medicine Program, Ottawa Hospital Research Institute.,Department of Medicine, The Ottawa Hospital and University of Ottawa.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa.,Centre for Neuromuscular Disease, University of Ottawa
| |
Collapse
|
83
|
Roles for RNA-binding proteins in development and disease. Brain Res 2016; 1647:1-8. [PMID: 26972534 DOI: 10.1016/j.brainres.2016.02.050] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
Abstract
RNA-binding protein activities are highly regulated through protein levels, intracellular localization, and post-translation modifications. During development, mRNA processing of specific gene sets is regulated through manipulation of functional RNA-binding protein activities. The impact of altered RNA-binding protein activities also affects human diseases in which there are either a gain-of-function or loss-of-function causes pathogenesis. We will discuss RNA-binding proteins and their normal developmental RNA metabolism and contrast how their function is disrupted in disease. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
|
84
|
Gudde AEEG, González-Barriga A, van den Broek WJAA, Wieringa B, Wansink DG. A low absolute number of expanded transcripts is involved in myotonic dystrophy type 1 manifestation in muscle. Hum Mol Genet 2016; 25:1648-62. [PMID: 26908607 PMCID: PMC4805313 DOI: 10.1093/hmg/ddw042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022] Open
Abstract
Muscular manifestation of myotonic dystrophy type 1 (DM1), a common inheritable degenerative multisystem disorder, is mainly caused by expression of RNA from a (CTG·CAG)n-expanded DM1 locus. Here, we report on comparative profiling of expression of normal and expanded endogenous or transgenic transcripts in skeletal muscle cells and biopsies from DM1 mouse models and patients in order to help us in understanding the role of this RNA-mediated toxicity. In tissue of HSALR mice, the most intensely used ‘muscle-only’ model in the DM1 field, RNA from the α-actin (CTG)250 transgene was at least 1000-fold more abundant than that from the Dmpk gene, or the DMPK gene in humans. Conversely, the DMPK transgene in another line, DM500/DMSXL mice, was expressed ∼10-fold lower than the endogenous gene. Temporal regulation of expanded RNA expression differed between models. Onset of expression occurred remarkably late in HSALR myoblasts during in vitro myogenesis whereas Dmpk or DMPK (trans)genes were expressed throughout proliferation and differentiation phases. Importantly, quantification of absolute transcript numbers revealed that normal and expanded Dmpk/DMPK transcripts in mouse models and DM1 patients are low-abundance RNA species. Northern blotting, reverse transcriptase–quantitative polymerase chain reaction, RNA-sequencing and fluorescent in situ hybridization analyses showed that they occur at an absolute number between one and a few dozen molecules per cell. Our findings refine the current RNA dominance theory for DM1 pathophysiology, as anomalous factor binding to expanded transcripts and formation of soluble or insoluble ribonucleoprotein aggregates must be nucleated by only few expanded DMPK transcripts and therefore be a small numbers game.
Collapse
Affiliation(s)
- Anke E E G Gudde
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Anchel González-Barriga
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Walther J A A van den Broek
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
85
|
Hypogonadism Associated with Cyp19a1 (Aromatase) Posttranscriptional Upregulation in Celf1 Knockout Mice. Mol Cell Biol 2015; 35:3244-53. [PMID: 26169831 DOI: 10.1128/mcb.00074-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 07/06/2015] [Indexed: 12/19/2022] Open
Abstract
CELF1 is a multifunctional RNA-binding protein that controls several aspects of RNA fate. The targeted disruption of the Celf1 gene in mice causes male infertility due to impaired spermiogenesis, the postmeiotic differentiation of male gametes. Here, we investigated the molecular reasons that underlie this testicular phenotype. By measuring sex hormone levels, we detected low concentrations of testosterone in Celf1-null mice. We investigated the effect of Celf1 disruption on the expression levels of steroidogenic enzyme genes, and we observed that Cyp19a1 was upregulated. Cyp19a1 encodes aromatase, which transforms testosterone into estradiol. Administration of testosterone or the aromatase inhibitor letrozole partly rescued the spermiogenesis defects, indicating that a lack of testosterone associated with excessive aromatase contributes to the testicular phenotype. In vivo and in vitro interaction assays demonstrated that CELF1 binds to Cyp19a1 mRNA, and reporter assays supported the conclusion that CELF1 directly represses Cyp19a1 translation. We conclude that CELF1 downregulates Cyp19a1 (Aromatase) posttranscriptionally to achieve high concentrations of testosterone compatible with spermiogenesis completion. We discuss the implications of these findings with respect to reproductive defects in men, including patients suffering from isolated hypogonadotropic hypogonadism and myotonic dystrophy type I.
Collapse
|
86
|
Mateos-Aierdi AJ, Goicoechea M, Aiastui A, Fernández-Torrón R, Garcia-Puga M, Matheu A, López de Munain A. Muscle wasting in myotonic dystrophies: a model of premature aging. Front Aging Neurosci 2015. [PMID: 26217220 PMCID: PMC4496580 DOI: 10.3389/fnagi.2015.00125] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1 or Steinert’s disease) and type 2 (DM2) are multisystem disorders of genetic origin. Progressive muscular weakness, atrophy and myotonia are the most prominent neuromuscular features of these diseases, while other clinical manifestations such as cardiomyopathy, insulin resistance and cataracts are also common. From a clinical perspective, most DM symptoms are interpreted as a result of an accelerated aging (cataracts, muscular weakness and atrophy, cognitive decline, metabolic dysfunction, etc.), including an increased risk of developing tumors. From this point of view, DM1 could be described as a progeroid syndrome since a notable age-dependent dysfunction of all systems occurs. The underlying molecular disorder in DM1 consists of the existence of a pathological (CTG) triplet expansion in the 3′ untranslated region (UTR) of the Dystrophia Myotonica Protein Kinase (DMPK) gene, whereas (CCTG)n repeats in the first intron of the Cellular Nucleic acid Binding Protein/Zinc Finger Protein 9(CNBP/ZNF9) gene cause DM2. The expansions are transcribed into (CUG)n and (CCUG)n-containing RNA, respectively, which form secondary structures and sequester RNA-binding proteins, such as the splicing factor muscleblind-like protein (MBNL), forming nuclear aggregates known as foci. Other splicing factors, such as CUGBP, are also disrupted, leading to a spliceopathy of a large number of downstream genes linked to the clinical features of these diseases. Skeletal muscle regeneration relies on muscle progenitor cells, known as satellite cells, which are activated after muscle damage, and which proliferate and differentiate to muscle cells, thus regenerating the damaged tissue. Satellite cell dysfunction seems to be a common feature of both age-dependent muscle degeneration (sarcopenia) and muscle wasting in DM and other muscle degenerative diseases. This review aims to describe the cellular, molecular and macrostructural processes involved in the muscular degeneration seen in DM patients, highlighting the similarities found with muscle aging.
Collapse
Affiliation(s)
- Alba Judith Mateos-Aierdi
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain
| | - Maria Goicoechea
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain
| | - Ana Aiastui
- CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Cell Culture Platform, Biodonostia Health Research Institute, San Sebastián Spain
| | - Roberto Fernández-Torrón
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Department of Neurology, Hospital Universitario Donostia, San Sebastián Spain
| | - Mikel Garcia-Puga
- Oncology Area, Biodonostia Health Research Institute San Sebastián, Spain
| | - Ander Matheu
- Oncology Area, Biodonostia Health Research Institute San Sebastián, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Department of Neurology, Hospital Universitario Donostia, San Sebastián Spain ; Department of Neuroscience, Universidad del País Vasco UPV-EHU San Sebastián, Spain
| |
Collapse
|
87
|
|
88
|
Reduction of toxic RNAs in myotonic dystrophies type 1 and type 2 by the RNA helicase p68/DDX5. Proc Natl Acad Sci U S A 2015; 112:8041-5. [PMID: 26080402 DOI: 10.1073/pnas.1422273112] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Myotonic dystrophies type 1 (DM1) and type 2 (DM2) are neuromuscular diseases, caused by accumulation of CUG and CCUG RNAs in toxic aggregates. Here we report that the increased stability of the mutant RNAs in both types of DM is caused by deficiency of RNA helicase p68. We have identified p68 by studying CCUG-binding proteins associated with degradation of the mutant CCUG repeats. Protein levels of p68 are reduced in DM1 and DM2 biopsied skeletal muscle. Delivery of p68 in DM1/2 cells causes degradation of the mutant RNAs, whereas delivery of p68 in skeletal muscle of DM1 mouse model reduces skeletal muscle myopathy and atrophy. Our study shows that correction of p68 may reduce toxicity of the mutant RNAs in DM1 and in DM2.
Collapse
|
89
|
MuSK frizzled-like domain is critical for mammalian neuromuscular junction formation and maintenance. J Neurosci 2015; 35:4926-41. [PMID: 25810523 DOI: 10.1523/jneurosci.3381-14.2015] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The muscle-specific kinase MuSK is one of the key molecules orchestrating neuromuscular junction (NMJ) formation. MuSK interacts with the Wnt morphogens, through its Frizzled-like domain (cysteine-rich domain [CRD]). Dysfunction of MuSK CRD in patients has been recently associated with the onset of myasthenia, common neuromuscular disorders mainly characterized by fatigable muscle weakness. However, the physiological role of Wnt-MuSK interaction in NMJ formation and function remains to be elucidated. Here, we demonstrate that the CRD deletion of MuSK in mice caused profound defects of both muscle prepatterning, the first step of NMJ formation, and synapse differentiation associated with a drastic deficit in AChR clusters and excessive growth of motor axons that bypass AChR clusters. Moreover, adult MuSKΔCRD mice developed signs of congenital myasthenia, including severe NMJs dismantlement, muscle weakness, and fatigability. We also report, for the first time, the beneficial effects of lithium chloride, a reversible inhibitor of the glycogen synthase kinase-3, that rescued NMJ defects in MuSKΔCRD mice and therefore constitutes a novel therapeutic reagent for the treatment of neuromuscular disorders linked to Wnt-MuSK signaling pathway deficiency. Together, our data reveal that MuSK CRD is critical for NMJ formation and plays an unsuspected role in NMJ maintenance in adulthood.
Collapse
|
90
|
Rahmati M, Taherabadi SJ, Mehrabi M. Decreased Activity in Neuropathic Pain Form and Gene Expression of Cyclin-Dependent Kinase5 and Glycogen Synthase Kinase-3 Beta in Soleus Muscle of Wistar Male Rats. IRANIAN RED CRESCENT MEDICAL JOURNAL 2015; 17:e23324. [PMID: 26290750 PMCID: PMC4537785 DOI: 10.5812/ircmj.23324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/18/2014] [Accepted: 03/25/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND The relationship between decreased activity/neuropathic pain and gene expression alterations in soleus muscle has remained elusive. OBJECTIVES In this experimental study, we investigated the effects of decreased activity in neuropathic pain form on Cyclin-Dependent Kinase 5 (CDK5) and Glycogen Synthase Kinase-3 β (GSK-3β) gene expression in soleus muscle of rats. MATERIALS AND METHODS Twelve male Wistar rats were randomly divided into three groups: (1) tight ligation of the L5 spinal nerve (SNL: n = 4); (2) sham surgery (Sham: n = 4), and (3) control (C: n = 4). The threshold to produce a withdrawal response to a mechanical and thermal stimulus was measured using von Frey filaments and radiation heat apparatus, respectively. Following 4 weeks after surgery, the left soleus muscle was removed and mRNA levels were determined by real-time Polymerase Chain Reaction (PCR). RESULTS Compared to control animals, L5 ligated animals developed mechanical and heat hypersensitivity during total period of study. Soleus muscle weight as well as CDK5 mRNA levels (less than ~ 0.4 fold) was decreased and GSK-3β mRNA levels (up to ~ 7 folds) increased in L5 ligated animals. CONCLUSIONS These results showed enhanced muscle atrophy processes following peripheral nerve damage and might provide a useful approach to study underlying muscle mechanisms associated with clinical neuropathic pain syndromes.
Collapse
Affiliation(s)
- Masoud Rahmati
- Department of Physical Education and Sport Sciences, Lorestan University, Khoram Abad, IR Iran
| | - Seyed Jalal Taherabadi
- Department of Physical Education and Sport Sciences, Lorestan University, Khoram Abad, IR Iran
| | - Mahmoud Mehrabi
- Department of Physical Education and Sport Sciences, Lorestan University, Khoram Abad, IR Iran
| |
Collapse
|
91
|
Yadava RS, Foff EP, Yu Q, Gladman JT, Kim YK, Bhatt KS, Thornton CA, Zheng TS, Mahadevan MS. TWEAK/Fn14, a pathway and novel therapeutic target in myotonic dystrophy. Hum Mol Genet 2015; 24:2035-48. [PMID: 25504044 PMCID: PMC4355029 DOI: 10.1093/hmg/ddu617] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/02/2014] [Accepted: 12/08/2014] [Indexed: 01/06/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.
Collapse
Affiliation(s)
| | - Erin P Foff
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | - Kirti S Bhatt
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Charles A Thornton
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Timothy S Zheng
- Department of Immunology, Biogen Idec, Cambridge, MA 02142, USA
| | | |
Collapse
|
92
|
Pettersson OJ, Aagaard L, Jensen TG, Damgaard CK. Molecular mechanisms in DM1 - a focus on foci. Nucleic Acids Res 2015; 43:2433-41. [PMID: 25605794 PMCID: PMC4344492 DOI: 10.1093/nar/gkv029] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/05/2015] [Accepted: 01/11/2015] [Indexed: 01/15/2023] Open
Abstract
Myotonic dystrophy type 1 is caused by abnormal expansion of a CTG-trinucleotide repeat in the gene encoding Dystrophia Myotonica Protein Kinase (DMPK), which in turn leads to global deregulation of gene expression in affected individuals. The transcribed mRNA contains a massive CUG-expansion in the 3' untranslated region (3'UTR) facilitating nucleation of several regulatory RNA-binding proteins, which are thus unable to perform their normal cellular function. These CUG-expanded mRNA-protein aggregates form distinct, primarily nuclear foci. In differentiated muscle cells, most of the CUG-expanded RNA remains in the nuclear compartment, while in dividing cells such as fibroblasts a considerable fraction of the mutant RNA reaches the cytoplasm, consistent with findings that both nuclear and cytoplasmic events are mis-regulated in DM1. Recent evidence suggests that the nuclear aggregates, or ribonuclear foci, are more dynamic than previously anticipated and regulated by several proteins, including RNA helicases. In this review, we focus on the homeostasis of DMPK mRNA foci and discuss how their dynamic regulation may affect disease-causing mechanisms in DM1.
Collapse
Affiliation(s)
- Olof Joakim Pettersson
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1240, DK-8000 Aarhus C, Denmark
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1240, DK-8000 Aarhus C, Denmark
| | - Thomas Gryesten Jensen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1240, DK-8000 Aarhus C, Denmark
| | - Christian Kroun Damgaard
- Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, Building 1130, DK-8000 Aarhus C, Denmark
| |
Collapse
|
93
|
Semaphorin3A-induced axonal transport mediated through phosphorylation of Axin-1 by GSK3β. Brain Res 2014; 1598:46-56. [PMID: 25528666 DOI: 10.1016/j.brainres.2014.12.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 11/22/2022]
Abstract
The establishment of neuronal polarity is necessary for proper neuronal wiring. Semaphorin3A (Sema3A), originally identified as a repulsive axon guidance molecule, exerts a wide variety of biological functions through signaling pathways including sequential phosphorylation of collapsin response mediator protein by cyclin-dependent kinase-5 (Cdk5) and glycogen synthase kinase-3β (GSK3β). Sema3A acts on its receptor neuropilin-1 to regulate axonal transport. To delineate mechanism by which Sema3A induces axonal transport, we investigate whether GSK3β is involved in mediating Sema3A-induced axonal transport. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione, an inhibitor of GSK3β, suppressed Sema3A-induced antero- and retrograde axonal transport. Introduction of either GSK3β mutants, GSK3β-L128A or K85M, suppressed Sema3A-induced axonal transport. On the other hand, introduction of GSK3β-R96A did not affect the Sema3A effect, suggesting that unprimed substrates are primarily involved in Sema3A-induced axonal transport. Overexpression of a partial fragment of frequently rearranged in advanced T-cell lymphomas 1 (FRATtide), which interferes the interaction between GSK3β and Axis inhibitor-1 (Axin-1), also suppressed Sema3A-induced transport. siRNA knockdown of Axin-1, an unprimed substrate of GSK3β, suppressed Sema3A-induced antero- and retrograde axonal transport. These results indicate that GSK3β and Axin-1 are involved in Sema3A-induced bidirectional axonal transport. This finding should provide a clue for understanding of mechanisms of a wide variety of biological activities of Sema3A.
Collapse
|
94
|
Ohsawa N, Koebis M, Mitsuhashi H, Nishino I, Ishiura S. ABLIM1 splicing is abnormal in skeletal muscle of patients with DM1 and regulated by MBNL, CELF and PTBP1. Genes Cells 2014; 20:121-34. [PMID: 25403273 DOI: 10.1111/gtc.12201] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/14/2014] [Indexed: 12/16/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is an RNA-mediated disorder characterized by muscle weakness, cardiac defects and multiple symptoms and is caused by expanded CTG repeats within the 3' untranslated region of the DMPK gene. In this study, we found abnormal splicing of actin-binding LIM protein 1 (ABLIM1) in skeletal muscles of patients with DM1 and a DM1 mouse model (HSA(LR) ). An exon 11 inclusion isoform is expressed in skeletal muscle and heart of non-DM1 individuals, but not in skeletal muscle of patients with DM1 or other adult human tissues. Moreover, we determined that ABLIM1 splicing is regulated by several splice factors, including MBNL family proteins, CELF1, 2 and 6, and PTBP1, using a cellular splicing assay. MBNL proteins promoted the inclusion of ABLIM1 exon 11, but other proteins and expanded CUG repeats repressed exon 11 of ABLIM1. This result is consistent with the hypothesis that MBNL proteins are trapped by expanded CUG repeats and inactivated in DM1 and that CELF1 is activated in DM1. However, activation of PTBP1 has not been reported in DM1. Our results suggest that the exon 11 inclusion isoform of ABLIM1 may have a muscle-specific function, and its abnormal splicing could be related to muscle symptoms of DM1.
Collapse
Affiliation(s)
- Natsumi Ohsawa
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo, 153-8902, Japan
| | | | | | | | | |
Collapse
|
95
|
Wojciechowska M, Taylor K, Sobczak K, Napierala M, Krzyzosiak WJ. Small molecule kinase inhibitors alleviate different molecular features of myotonic dystrophy type 1. RNA Biol 2014; 11:742-54. [PMID: 24824895 PMCID: PMC4156505 DOI: 10.4161/rna.28799] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Expandable (CTG)n repeats in the 3′ UTR of the DMPK gene are a cause of myotonic dystrophy type 1 (DM1), which leads to a toxic RNA gain-of-function disease. Mutant RNAs with expanded CUG repeats are retained in the nucleus and aggregate in discrete inclusions. These foci sequester splicing factors of the MBNL family and trigger upregulation of the CUGBP family of proteins resulting in the mis-splicing of their target transcripts. To date, many efforts to develop novel therapeutic strategies have been focused on disrupting the toxic nuclear foci and correcting aberrant alternative splicing via targeting mutant CUG repeats RNA; however, no effective treatment for DM1 is currently available. Herein, we present results of culturing of human DM1 myoblasts and fibroblasts with two small-molecule ATP-binding site-specific kinase inhibitors, C16 and C51, which resulted in the alleviation of the dominant-negative effects of CUG repeat expansion. Reversal of the DM1 molecular phenotype includes a reduction of the size and number of foci containing expanded CUG repeat transcripts, decreased steady-state levels of CUGBP1 protein, and consequent improvement of the aberrant alternative splicing of several pre-mRNAs misregulated in DM1.
Collapse
Affiliation(s)
- Marzena Wojciechowska
- Department of Molecular Biomedicine; Institute of Bioorganic Chemistry; Polish Academy of Sciences; Noskowskiego; Poznan, Poland
| | - Katarzyna Taylor
- Department of Gene Expression; Institute of Molecular Biology and Biotechnology; Adam Mickiewicz University; Umultowska 89; Poznan, Poland
| | - Krzysztof Sobczak
- Department of Gene Expression; Institute of Molecular Biology and Biotechnology; Adam Mickiewicz University; Umultowska 89; Poznan, Poland
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics and UAB Stem Cell Institute; University of Alabama at Birmingham; Birmingham, AL USA
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine; Institute of Bioorganic Chemistry; Polish Academy of Sciences; Noskowskiego; Poznan, Poland
| |
Collapse
|
96
|
Wei C, Jones K, Timchenko NA, Timchenko L. GSK3β is a new therapeutic target for myotonic dystrophy type 1. Rare Dis 2013; 1:e26555. [PMID: 25003008 PMCID: PMC3927489 DOI: 10.4161/rdis.26555] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/16/2013] [Accepted: 09/20/2013] [Indexed: 12/31/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1), an incurable, neuromuscular disease, is caused by the expansion of CTG repeats within the 3′ UTR of DMPK on chromosome 19q. In DM1 patients, mutant DMPK transcripts deregulate RNA metabolism by altering CUG RNA-binding proteins. Several approaches have been proposed for DM1 therapy focused on specific degradation of the mutant CUG repeats or on correction of RNA-binding proteins, affected by CUG repeats. One such protein is CUG RNA-binding protein (CUGBP1). The ability of CUGBP1 to increase or inhibit translation depends on phosphorylation at Ser302, which is mediated by cyclin D3-CDK4. The mutant CUG repeats increase the levels of CUGBP1 protein and inhibit Ser302 phosphorylation, leading to the accumulation of CUGBP1 isoforms that repress translation (i.e., CUGBP1REP). Elevation of CUGBP1REP in DM1 is caused by increased GSK3β kinase, which reduces the cyclin D3-CDK4 pathway and subsequent phosphorylation of CUGBP1 at Ser302. In this review, we discuss our recent discovery showing that correction of GSK3β activity in the DM1 mouse model (i.e., HSALR mice) reduces DM1 muscle pathology. These findings demonstrate that GSK3β is a novel therapeutic target for treating DM1.
Collapse
Affiliation(s)
- Christina Wei
- Department of Molecular Physiology and Biophysics; Baylor College of Medicine; Houston, TX USA
| | - Karlie Jones
- Department of Molecular Physiology and Biophysics; Baylor College of Medicine; Houston, TX USA
| | - Nikolai A Timchenko
- Pathology and Immunology; Huffington Center on Aging; Baylor College of Medicine; Houston, TX USA
| | - Lubov Timchenko
- Department of Molecular Physiology and Biophysics; Baylor College of Medicine; Houston, TX USA
| |
Collapse
|
97
|
Richards RI, Samaraweera SE, van Eyk CL, O'Keefe LV, Suter CM. RNA pathogenesis via Toll-like receptor-activated inflammation in expanded repeat neurodegenerative diseases. Front Mol Neurosci 2013; 6:25. [PMID: 24046729 PMCID: PMC3763583 DOI: 10.3389/fnmol.2013.00025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 08/14/2013] [Indexed: 12/13/2022] Open
Abstract
Previously, we hypothesized that an RNA-based pathogenic pathway has a causal role in the dominantly inherited unstable expanded repeat neurodegenerative diseases. In support of this hypothesis we, and others, have characterized rCAG.rCUG100 repeat double-strand RNA (dsRNA) as a previously unidentified agent capable of causing pathogenesis in a Drosophila model of neurodegenerative disease. Dicer, Toll, and autophagy pathways have distinct roles in this Drosophila dsRNA pathology. Dicer dependence is accompanied by cleavage of rCAG.rCUG100 repeat dsRNA down to r(CAG)7 21-mers. Among the “molecular hallmarks” of this pathway that have been identified in Drosophila, some [i.e., r(CAG)7 and elevated tumor necrosis factor] correlate with observations in affected people (e.g., Huntington’s disease and amyotrophic lateral sclerosis) or in related animal models (i.e., autophagy). The Toll pathway is activated in the presence of repeat-containing dsRNA and toxicity is also dependent on this pathway. How might the endogenously expressed dsRNA mediate Toll-dependent toxicity in neuronal cells? Endogenous RNAs are normally shielded from Toll pathway activation as part of the mechanism to distinguish “self” from “non-self” RNAs. This typically involves post-transcriptional modification of the RNA. Therefore, it is likely that rCAG.rCUG100 repeat dsRNA has a characteristic property that interferes with or evades this normal mechanism of shielding. We predict that repeat expansion leads to an alteration in RNA structure and/or form that perturbs RNA modification, causing the unshielded repeat RNA (in the form of its Dicer-cleaved products) to be recognized by Toll-like receptors (TLRs), with consequent activation of the Toll pathway leading to loss of cell function and then ultimately cell death. We hypothesize that the proximal cause of expanded repeat neurodegenerative diseases is the TLR recognition (and resultant innate inflammatory response) of repeat RNA as “non-self” due to their paucity of “self” modification.
Collapse
Affiliation(s)
- Robert I Richards
- Discipline of Genetics and Centre for Molecular Pathology, School of Molecular and Biomedical Science, The University of Adelaide Adelaide, SA, Australia
| | | | | | | | | |
Collapse
|
98
|
Reexpression of pyruvate kinase M2 in type 1 myofibers correlates with altered glucose metabolism in myotonic dystrophy. Proc Natl Acad Sci U S A 2013; 110:13570-5. [PMID: 23901116 DOI: 10.1073/pnas.1308806110] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is caused by expansion of CTG repeats in the 3' UTR of the DMPK gene. Expression of CUG expansion (CUG(exp)) RNA produces a toxic gain of function by disrupting the functions of RNA splicing factors, such as MBNL1 and CELF1, leading to splicing changes associated with clinical abnormalities. Progressive skeletal muscle weakness and wasting is one of the most prominent clinical features in DM1; however, the underlying mechanisms remain unclear. Here we report that the embryonic M2 isoform of pyruvate kinase (PKM2), a key enzyme contributing to the Warburg effect in cancer, is significantly induced in DM1 tissue and mouse models owing to aberrant splicing. Expression of PKM2 in DM1 skeletal muscle is restricted to the type 1 fibers, which are particularly susceptible to wasting in DM1. Using antisense oligonucleotides to shift PKM splicing toward increased PKM2 expression, we observed increased glucose consumption with reduced oxidative metabolism in cell culture and increased respiratory exchange ratio in mice, suggesting defects in energy metabolism conferred by PKM2 expression. We propose that PKM2 expression induces changes in type 1 fibers associated with muscle atrophy and muscle weakness in DM1.
Collapse
|
99
|
Michalova E, Vojtesek B, Hrstka R. Impaired pre-mRNA processing and altered architecture of 3' untranslated regions contribute to the development of human disorders. Int J Mol Sci 2013; 14:15681-94. [PMID: 23896598 PMCID: PMC3759880 DOI: 10.3390/ijms140815681] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 06/21/2013] [Accepted: 06/24/2013] [Indexed: 11/16/2022] Open
Abstract
The biological fate of each mRNA and consequently, the protein to be synthesised, is highly dependent on the nature of the 3' untranslated region. Despite its non-coding character, the 3' UTR may affect the final mRNA stability, the localisation, the export from the nucleus and the translation efficiency. The conserved regulatory sequences within 3' UTRs and the specific elements binding to them enable gene expression control at the posttranscriptional level and all these processes reflect the actual state of the cell including proliferation, differentiation, cellular stress or tumourigenesis. Through this article, we briefly outline how the alterations in the establishment and final architecture of 3' UTRs may contribute to the development of various disorders in humans.
Collapse
Affiliation(s)
- Eva Michalova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 656 53, Czech Republic; E-Mails: (E.M.); (B.V.)
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 656 53, Czech Republic; E-Mails: (E.M.); (B.V.)
| | - Roman Hrstka
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno 656 53, Czech Republic; E-Mails: (E.M.); (B.V.)
| |
Collapse
|
100
|
Molecular mechanisms of muscle atrophy in myotonic dystrophies. Int J Biochem Cell Biol 2013; 45:2280-7. [PMID: 23796888 DOI: 10.1016/j.biocel.2013.06.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 02/01/2023]
Abstract
Myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2) are multisystemic diseases that primarily affect skeletal muscle, causing myotonia, muscle atrophy, and muscle weakness. DM1 and DM2 pathologies are caused by expansion of CTG and CCTG repeats in non-coding regions of the genes encoding myotonic dystrophy protein kinase (DMPK) and zinc finger protein 9 (ZNF9) respectively. These expansions cause DM pathologies through accumulation of mutant RNAs that alter RNA metabolism in patients' tissues by targeting RNA-binding proteins such as CUG-binding protein 1 (CUGBP1) and Muscle blind-like protein 1 (MBNL1). Despite overwhelming evidence showing the critical role of RNA-binding proteins in DM1 and DM2 pathologies, the downstream pathways by which these RNA-binding proteins cause muscle wasting and muscle weakness are not well understood. This review discusses the molecular pathways by which DM1 and DM2 mutations might cause muscle atrophy and describes progress toward the development of therapeutic interventions for muscle wasting and weakness in DM1 and DM2. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
|