1
|
Jennings K, Lindquist D, Poonia A, Schoser B, Schneider-Gold C, Timchenko NA, Timchenko L. The role of CNBP in brain atrophy and its targeting in myotonic dystrophy type 2. Hum Mol Genet 2025; 34:512-522. [PMID: 39807631 DOI: 10.1093/hmg/ddaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Myotonic Dystrophy type 2 (DM2) is a multisystem disease affecting many tissues, including skeletal muscle, heart, and brain. DM2 is caused by unstable expansion of CCTG repeats in an intron 1 of a gene coding for cellular nuclear binding protein (CNBP). The expanded CCTG repeats cause DM2 pathology due to the accumulation of RNA CCUG repeats, which affect RNA processing in patients' cells. We have previously shown that mutant CCUG repeats reduce CNBP protein in DM2 patients. Reducing Cnbp in Cnbp KO mouse model causes late skeletal muscle atrophy. In this study, we examined if the reduction of Cnbp affects the Central Nervous System (CNS). MRI and DTI analyses showed that total brain volume and grey matter are reduced in Cnbp KO mice, while mean, radial and axonal brain diffusivity is increased. The morphological changes in the brains of Cnbp KO mice are accompanied by reduced stereotypic behavior, anxiety and neuromotor defects. These findings suggest that the reduction of CNBP contributes to CNS pathology in DM2. Since CNBP stability is regulated by pAMPK-dependent phosphorylation, we examined protein levels of pAMPK in DM2 cells and found that the active pAMPK is reduced in DM2. Interaction of CNBP with pAMPK and stability of CNBP protein are also decreased in DM2. Our data show that a small molecule AMPK activator A769662 corrects CNBP stability and normalizes CNBP targets in DM2 fibroblasts. Thus, activators of AMPK could potentially be developed as therapeutics to correct CNBP and reduce muscle and brain atrophies in DM2.
Collapse
Affiliation(s)
- Katherine Jennings
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Diana Lindquist
- Imaging Research Center, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
- Departments of Radiology, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
| | - Ankita Poonia
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Benedikt Schoser
- Department of Neurology, Friedrich-Baur-Institute, LMU Clinics, Ziemssenstr, 1 Munich 80336, Germany
| | - Christiane Schneider-Gold
- Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Gudrinstr. 56, Bochum 44791, Germany
| | - Nikolai A Timchenko
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
- Department of Surgery, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
| | - Lubov Timchenko
- Division of Neurology, Cincinnati Children's Hospital, 3333 Burnet Ave, Cincinnati, OH 45229, United States
- Pediatrics, University of Cincinnati, 2600 Clifton Ave, Cincinnati, OH 45221, United States
| |
Collapse
|
2
|
Rösing S, Ullrich F, Meisterfeld S, Schmidt F, Mlitzko L, Croon M, Nattrass RG, Eberl N, Mahlberg J, Schlee M, Wieland A, Simon P, Hilbig D, Reuner U, Rapp A, Bremser J, Mirtschink P, Drukewitz S, Zillinger T, Beissert S, Paeschke K, Hartmann G, Trifunovic A, Bartok E, Günther C. Chronic endoplasmic reticulum stress in myotonic dystrophy type 2 promotes autoimmunity via mitochondrial DNA release. Nat Commun 2024; 15:1534. [PMID: 38378748 PMCID: PMC10879130 DOI: 10.1038/s41467-024-45535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Myotonic dystrophy type 2 (DM2) is a tetranucleotide CCTG repeat expansion disease associated with an increased prevalence of autoimmunity. Here, we identified an elevated type I interferon (IFN) signature in peripheral blood mononuclear cells and primary fibroblasts of DM2 patients as a trigger of chronic immune stimulation. Although RNA-repeat accumulation was prevalent in the cytosol of DM2-patient fibroblasts, type-I IFN release did not depend on innate RNA immune sensors but rather the DNA sensor cGAS and the prevalence of mitochondrial DNA (mtDNA) in the cytoplasm. Sublethal mtDNA release was promoted by a chronic activation of the ATF6 branch of the unfolded protein response (UPR) in reaction to RNA-repeat accumulation and non-AUG translated tetrapeptide expansion proteins. ATF6-dependent mtDNA release and resulting cGAS/STING activation could also be recapitulated in human THP-1 monocytes exposed to chronic endoplasmic reticulum (ER) stress. Altogether, our study demonstrates a novel mechanism by which large repeat expansions cause chronic endoplasmic reticulum stress and associated mtDNA leakage. This mtDNA is, in turn, sensed by the cGAS/STING pathway and induces a type-I IFN response predisposing to autoimmunity. Elucidating this pathway reveals new potential therapeutic targets for autoimmune disorders associated with repeat expansion diseases.
Collapse
Affiliation(s)
- Sarah Rösing
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Fabian Ullrich
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Susann Meisterfeld
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Franziska Schmidt
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Laura Mlitzko
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Marijana Croon
- Institute for Mitochondrial Diseases and Aging, Faculty of Medicine, CECAD Research Center, 50931, Cologne, Germany
| | - Ryan G Nattrass
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Nadia Eberl
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Julia Mahlberg
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Anja Wieland
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Philipp Simon
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127, Bonn, Germany
| | - Daniel Hilbig
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127, Bonn, Germany
| | - Ulrike Reuner
- Department of Neurology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Alexander Rapp
- Department of Biology, Cell biology and Epigenetic, Technical University of Darmstadt, Darmstadt, Germany
| | - Julia Bremser
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, 01307, Dresden, Germany
| | - Stephan Drukewitz
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Partner Site Dresden, Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Stefan Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany
| | - Katrin Paeschke
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, 53127, Bonn, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Aleksandra Trifunovic
- Institute for Mitochondrial Diseases and Aging, Faculty of Medicine, CECAD Research Center, 50931, Cologne, Germany
| | - Eva Bartok
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, 53127, Bonn, Germany
- Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Claudia Günther
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307, Dresden, Germany.
| |
Collapse
|
3
|
Peric S, Zlatar J, Nikolic L, Ivanovic V, Pesovic J, Petrovic Djordjevic I, Sreckovic S, Savic-Pavicevic D, Meola G, Rakocevic-Stojanovic V. Autoimmune Diseases in Patients With Myotonic Dystrophy Type 2. Front Neurol 2022; 13:932883. [PMID: 35923829 PMCID: PMC9341519 DOI: 10.3389/fneur.2022.932883] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Myotonic dystrophy type 2 (DM2) is a rare autosomal dominant multisystemic disease with highly variable clinical presentation. Several case reports and one cohort study suggested a significant association between DM2 and autoimmune diseases (AIDs). Aim The aim of this study is to analyze the frequency and type of AIDs in patients with DM2 from the Serbian DM registry. Patients and Methods A total of 131 patients with DM2 from 108 families were included, [62.6% women, mean age at DM2 onset 40.4 (with standard deviation 13) years, age at entering the registry 52 (12.8) years, and age at analysis 58.4 (12.8) years]. Data were obtained from Akhenaten, the Serbian registry for DM, and through the hospital electronic data system. Results Upon entering the registry, 35 (26.7%) of the 131 patients with DM2 had AIDs including Hashimoto thyroiditis (18.1%), rheumatoid arthritis, diabetes mellitus type 1, systemic lupus, Sjogren's disease, localized scleroderma, psoriasis, celiac disease, Graves's disease, neuromyelitis optica, myasthenia gravis, and Guillain-Barre syndrome. At the time of data analysis, one additional patient developed new AIDs, so eventually, 36 (28.8%) of 125 DM2 survivors had AIDs. Antinuclear antibodies (ANAs) were found in 14 (10.7%) of 63 tested patients, including 12 without defined corresponding AID (all in low titers, 1:40 to 1:160). Antineutrophil cytoplasmic antibodies (ANCAs) were negative in all 50 tested cases. The percentage of women was significantly higher among patients with AIDs (82.9% vs. 55.2%, p <0.01). Conclusion AIDs were present in as high as 30% of the patients with DM2. Thus, screening for AIDs in DM2 seems reasonable. Presence of AIDs and/or ANAs may lead to under-diagnosis of DM2.
Collapse
Affiliation(s)
- Stojan Peric
- Neurology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- *Correspondence: Stojan Peric ; orcid.org/0000-0002-2979-556X
| | - Jelena Zlatar
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Luka Nikolic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vukan Ivanovic
- Neurology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | - Jovan Pesovic
- Center for Human Molecular Genetics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ivana Petrovic Djordjevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Cardiology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | - Svetlana Sreckovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Anaesthesiology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
| | - Dusanka Savic-Pavicevic
- Center for Human Molecular Genetics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Giovanni Meola
- Department of Neurorehabilitation Sciences, Casa Di Cura del Policlinico, Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Vidosava Rakocevic-Stojanovic
- Neurology Clinic, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Characterisation of Non-Pathogenic Premutation-Range Myotonic Dystrophy Type 2 Alleles. J Clin Med 2021; 10:jcm10173934. [PMID: 34501382 PMCID: PMC8432210 DOI: 10.3390/jcm10173934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022] Open
Abstract
Myotonic dystrophy type 2 (DM2) is caused by expansion of a (CCTG)n repeat in the cellular retroviral nucleic acid-binding protein (CNBP) gene. The sequence of the repeat is most commonly interrupted and is stably inherited in the general population. Although expanded alleles, premutation range and, in rare cases, also non-disease associated alleles containing uninterrupted CCTG tracts have been described, the threshold between these categories is poorly characterised. Here, we describe four families with members reporting neuromuscular complaints, in whom we identified altogether nine ambiguous CNBP alleles containing uninterrupted CCTG repeats in the range between 32 and 42 repeats. While these grey-zone alleles are most likely not pathogenic themselves, since other pathogenic mutations were identified and particular family structures did not support their pathogenic role, they were found to be unstable during intergenerational transmission. On the other hand, there was no observable general microsatellite instability in the genome of the carriers of these alleles. Our results further refine the division of CNBP CCTG repeat alleles into two major groups, i.e., interrupted and uninterrupted alleles. Both interrupted and uninterrupted alleles with up to approximately 30 CCTG repeats were shown to be generally stable during intergenerational transmission, while intergenerational as well as somatic instability seems to gradually increase in uninterrupted alleles with tract length growing above this threshold.
Collapse
|
5
|
Nishimura AL, Arias N. Synaptopathy Mechanisms in ALS Caused by C9orf72 Repeat Expansion. Front Cell Neurosci 2021; 15:660693. [PMID: 34140881 PMCID: PMC8203826 DOI: 10.3389/fncel.2021.660693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease caused by degeneration of motor neurons (MNs). ALS pathogenic features include accumulation of misfolded proteins, glutamate excitotoxicity, mitochondrial dysfunction at distal axon terminals, and neuronal cytoskeleton changes. Synergies between loss of C9orf72 functions and gain of function by toxic effects of repeat expansions also contribute to C9orf72-mediated pathogenesis. However, the impact of haploinsufficiency of C9orf72 on neurons and in synaptic functions requires further examination. As the motor neurons degenerate, the disease symptoms will lead to neurotransmission deficiencies in the brain, spinal cord, and neuromuscular junction. Altered neuronal excitability, synaptic morphological changes, and C9orf72 protein and DPR localization at the synapses, suggest a potential involvement of C9orf72 at synapses. In this review article, we provide a conceptual framework for assessing the putative involvement of C9orf72 as a synaptopathy, and we explore the underlying and common disease mechanisms with other neurodegenerative diseases. Finally, we reflect on the major challenges of understanding C9orf72-ALS as a synaptopathy focusing on integrating mitochondrial and neuronal cytoskeleton degeneration as biomarkers and potential targets to treat ALS neurodegeneration.
Collapse
Affiliation(s)
- Agnes L Nishimura
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, UK Dementia Research Institute, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
6
|
Braems E, Swinnen B, Van Den Bosch L. C9orf72 loss-of-function: a trivial, stand-alone or additive mechanism in C9 ALS/FTD? Acta Neuropathol 2020; 140:625-643. [PMID: 32876811 PMCID: PMC7547039 DOI: 10.1007/s00401-020-02214-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/28/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
A repeat expansion in C9orf72 is responsible for the characteristic neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) in a still unresolved manner. Proposed mechanisms involve gain-of-functions, comprising RNA and protein toxicity, and loss-of-function of the C9orf72 gene. Their exact contribution is still inconclusive and reports regarding loss-of-function are rather inconsistent. Here, we review the function of the C9orf72 protein and its relevance in disease. We explore the potential link between reduced C9orf72 levels and disease phenotypes in postmortem, in vitro, and in vivo models. Moreover, the significance of loss-of-function in other non-coding repeat expansion diseases is used to clarify its contribution in C9orf72 ALS/FTD. In conclusion, with evidence pointing to a multiple-hit model, loss-of-function on itself seems to be insufficient to cause neurodegeneration in C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Elke Braems
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium
- Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N4, Herestraat 49, PB 602, 3000, Leuven, Belgium
| | - Bart Swinnen
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium
- Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N4, Herestraat 49, PB 602, 3000, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000, Leuven, Belgium.
- Laboratory of Neurobiology, Experimental Neurology, Center for Brain and Disease Research, VIB, Campus Gasthuisberg, O&N4, Herestraat 49, PB 602, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
An Overview of Alternative Splicing Defects Implicated in Myotonic Dystrophy Type I. Genes (Basel) 2020; 11:genes11091109. [PMID: 32971903 PMCID: PMC7564762 DOI: 10.3390/genes11091109] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 01/02/2023] Open
Abstract
Myotonic dystrophy type I (DM1) is the most common form of adult muscular dystrophy, caused by expansion of a CTG triplet repeat in the 3′ untranslated region (3′UTR) of the myotonic dystrophy protein kinase (DMPK) gene. The pathological CTG repeats result in protein trapping by expanded transcripts, a decreased DMPK translation and the disruption of the chromatin structure, affecting neighboring genes expression. The muscleblind-like (MBNL) and CUG-BP and ETR-3-like factors (CELF) are two families of tissue-specific regulators of developmentally programmed alternative splicing that act as antagonist regulators of several pre-mRNA targets, including troponin 2 (TNNT2), insulin receptor (INSR), chloride channel 1 (CLCN1) and MBNL2. Sequestration of MBNL proteins and up-regulation of CELF1 are key to DM1 pathology, inducing a spliceopathy that leads to a developmental remodelling of the transcriptome due to an adult-to-foetal splicing switch, which results in the loss of cell function and viability. Moreover, recent studies indicate that additional pathogenic mechanisms may also contribute to disease pathology, including a misregulation of cellular mRNA translation, localization and stability. This review focuses on the cause and effects of MBNL and CELF1 deregulation in DM1, describing the molecular mechanisms underlying alternative splicing misregulation for a deeper understanding of DM1 complexity. To contribute to this analysis, we have prepared a comprehensive list of transcript alterations involved in DM1 pathogenesis, as well as other deregulated mRNA processing pathways implications.
Collapse
|
8
|
Swinnen B, Robberecht W, Van Den Bosch L. RNA toxicity in non-coding repeat expansion disorders. EMBO J 2020; 39:e101112. [PMID: 31721251 PMCID: PMC6939197 DOI: 10.15252/embj.2018101112] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Several neurodegenerative disorders like amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia (SCA) are caused by non-coding nucleotide repeat expansions. Different pathogenic mechanisms may underlie these non-coding repeat expansion disorders. While gain-of-function mechanisms, such as toxicity associated with expression of repeat RNA or toxicity associated with repeat-associated non-ATG (RAN) products, are most frequently connected with these disorders, loss-of-function mechanisms have also been implicated. We review the different pathways that have been linked to non-coding repeat expansion disorders such as C9ORF72-linked ALS/frontotemporal dementia (FTD), myotonic dystrophy, fragile X tremor/ataxia syndrome (FXTAS), SCA, and Huntington's disease-like 2. We discuss modes of RNA toxicity focusing on the identity and the interacting partners of the toxic RNA species. Using the C9ORF72 ALS/FTD paradigm, we further explore the efforts and different methods used to disentangle RNA vs. RAN toxicity. Overall, we conclude that there is ample evidence for a role of RNA toxicity in non-coding repeat expansion diseases.
Collapse
Affiliation(s)
- Bart Swinnen
- Department of NeurosciencesExperimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Wim Robberecht
- Department of NeurosciencesExperimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Ludo Van Den Bosch
- Department of NeurosciencesExperimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| |
Collapse
|
9
|
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
|
10
|
Sellier C, Cerro-Herreros E, Blatter M, Freyermuth F, Gaucherot A, Ruffenach F, Sarkar P, Puymirat J, Udd B, Day JW, Meola G, Bassez G, Fujimura H, Takahashi MP, Schoser B, Furling D, Artero R, Allain FHT, Llamusi B, Charlet-Berguerand N. rbFOX1/MBNL1 competition for CCUG RNA repeats binding contributes to myotonic dystrophy type 1/type 2 differences. Nat Commun 2018; 9:2009. [PMID: 29789616 PMCID: PMC5964235 DOI: 10.1038/s41467-018-04370-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 04/26/2018] [Indexed: 12/30/2022] Open
Abstract
Myotonic dystrophy type 1 and type 2 (DM1, DM2) are caused by expansions of CTG and CCTG repeats, respectively. RNAs containing expanded CUG or CCUG repeats interfere with the metabolism of other RNAs through titration of the Muscleblind-like (MBNL) RNA binding proteins. DM2 follows a more favorable clinical course than DM1, suggesting that specific modifiers may modulate DM severity. Here, we report that the rbFOX1 RNA binding protein binds to expanded CCUG RNA repeats, but not to expanded CUG RNA repeats. Interestingly, rbFOX1 competes with MBNL1 for binding to CCUG expanded repeats and overexpression of rbFOX1 partly releases MBNL1 from sequestration within CCUG RNA foci in DM2 muscle cells. Furthermore, expression of rbFOX1 corrects alternative splicing alterations and rescues muscle atrophy, climbing and flying defects caused by expression of expanded CCUG repeats in a Drosophila model of DM2.
Collapse
Affiliation(s)
- Chantal Sellier
- IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, 67404, Illkirch, France
| | - Estefanía Cerro-Herreros
- Translational Genomics Group, Interdisciplinary Research Structure for Biotechnology and Biomedicine BIOTECMED, University of Valencia, 46010, Valencia, Spain
- INCLIVA Health Research Institute, 46010, Valencia, Spain
| | - Markus Blatter
- Institute for Molecular Biology and Biophysics, Swiss Federal Institute of Technology (ETH) Zurich, 8092, Zurich, Switzerland
| | - Fernande Freyermuth
- IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, 67404, Illkirch, France
| | - Angeline Gaucherot
- IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, 67404, Illkirch, France
| | - Frank Ruffenach
- IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, 67404, Illkirch, France
| | - Partha Sarkar
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jack Puymirat
- Human Genetics Research Unit, Laval University, CHUQ, Ste-Foy, Quebec, QC G1V 4G2, Canada
| | - Bjarne Udd
- Neuromuscular Research Center, Tampere University Hospital, 33521, Tampere, Finland
- Department of Medical Genetics, Folkhälsan Institute of Genetics, Helsinki University, 00290, Helsinki, Finland
- Department of Neurology, Vasa Central Hospital, 65130, Vaasa, Finland
| | - John W Day
- Department of Neurology, Stanford University, San Francisco, CA, 94305, USA
| | - Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, 20097, Milan, Italy
- Neurology Unit, IRCCS Policlinico San Donato, San Donato Milanese, 20097, Milan, Italy
| | - Guillaume Bassez
- Sorbonne Université, Inserm, Association Institut de Myologie, Center of Research in Myology, 75013, Paris, France
| | - Harutoshi Fujimura
- Department of Neurology, Toneyama National Hospital, Toyonaka, 560-0045, Japan
| | - Masanori P Takahashi
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, Ludwig Maximilian University, 80539, Munich, Germany
| | - Denis Furling
- Sorbonne Université, Inserm, Association Institut de Myologie, Center of Research in Myology, 75013, Paris, France
| | - Ruben Artero
- Translational Genomics Group, Interdisciplinary Research Structure for Biotechnology and Biomedicine BIOTECMED, University of Valencia, 46010, Valencia, Spain
- INCLIVA Health Research Institute, 46010, Valencia, Spain
| | - Frédéric H T Allain
- Institute for Molecular Biology and Biophysics, Swiss Federal Institute of Technology (ETH) Zurich, 8092, Zurich, Switzerland
| | - Beatriz Llamusi
- Translational Genomics Group, Interdisciplinary Research Structure for Biotechnology and Biomedicine BIOTECMED, University of Valencia, 46010, Valencia, Spain.
- INCLIVA Health Research Institute, 46010, Valencia, Spain.
| | - Nicolas Charlet-Berguerand
- IGBMC, INSERM U964, CNRS UMR7104, University of Strasbourg, 67404, Illkirch, France.
- UMR7104, Centre National de la Recherche Scientifique, 67404, Illkirch, France.
- Institut National de la Santé et de la Recherche Médicale, U964, 67404, Illkirch, France.
- Université de Strasbourg, 67404, Illkirch, France.
| |
Collapse
|
11
|
Santoro M, Fontana L, Maiorca F, Centofanti F, Massa R, Silvestri G, Novelli G, Botta A. Expanded [CCTG]n repetitions are not associated with abnormal methylation at the CNBP locus in myotonic dystrophy type 2 (DM2) patients. Biochim Biophys Acta Mol Basis Dis 2018; 1864:917-924. [DOI: 10.1016/j.bbadis.2017.12.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 12/21/2017] [Accepted: 12/28/2017] [Indexed: 01/10/2023]
|
12
|
Yenigun VB, Sirito M, Amcheslavky A, Czernuszewicz T, Colonques-Bellmunt J, García-Alcover I, Wojciechowska M, Bolduc C, Chen Z, López Castel A, Krahe R, Bergmann A. (CCUG) n RNA toxicity in a Drosophila model of myotonic dystrophy type 2 (DM2) activates apoptosis. Dis Model Mech 2017. [PMID: 28623239 PMCID: PMC5560059 DOI: 10.1242/dmm.026179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The myotonic dystrophies are prototypic toxic RNA gain-of-function diseases. Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are caused by different unstable, noncoding microsatellite repeat expansions – (CTG)DM1 in DMPK and (CCTG)DM2 in CNBP. Although transcription of mutant repeats into (CUG)DM1 or (CCUG)DM2 appears to be necessary and sufficient to cause disease, their pathomechanisms remain incompletely understood. To study the mechanisms of (CCUG)DM2 toxicity and develop a convenient model for drug screening, we generated a transgenic DM2 model in the fruit fly Drosophila melanogaster with (CCUG)n repeats of variable length (n=16 and 106). Expression of noncoding (CCUG)106, but not (CCUG)16, in muscle and retinal cells led to the formation of ribonuclear foci and mis-splicing of genes implicated in DM pathology. Mis-splicing could be rescued by co-expression of human MBNL1, but not by CUGBP1 (CELF1) complementation. Flies with (CCUG)106 displayed strong disruption of external eye morphology and of the underlying retina. Furthermore, expression of (CCUG)106 in developing retinae caused a strong apoptotic response. Inhibition of apoptosis rescued the retinal disruption in (CCUG)106 flies. Finally, we tested two chemical compounds that have shown therapeutic potential in DM1 models. Whereas treatment of (CCUG)106 flies with pentamidine had no effect, treatment with a PKR inhibitor blocked both the formation of RNA foci and apoptosis in retinae of (CCUG)106 flies. Our data indicate that expression of expanded (CCUG)DM2 repeats is toxic, causing inappropriate cell death in affected fly eyes. Our Drosophila DM2 model might provide a convenient tool for in vivo drug screening. Summary: A Drosophila model of myotonic dystrophy type 2 (DM2) recapitulates several features of the human disease, identifies apoptosis as a contributing factor to DM2, and is likely to provide a convenient tool for drug screening.
Collapse
Affiliation(s)
- Vildan Betul Yenigun
- Department of Biochemistry & Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Programs in Genes & Development, University of Texas Graduate School in Biomedical Sciences at Houston, Houston, TX, USA
| | - Mario Sirito
- Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alla Amcheslavky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Tomek Czernuszewicz
- Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Marzena Wojciechowska
- Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clare Bolduc
- Department of Biochemistry & Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihong Chen
- Department of Biochemistry & Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Ralf Krahe
- Departments of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA .,Graduate Programs in Genes & Development, University of Texas Graduate School in Biomedical Sciences at Houston, Houston, TX, USA.,Graduate Programs in Human & Molecular Genetics, University of Texas Graduate School in Biomedical Sciences at Houston, Houston, Texas, USA
| | - Andreas Bergmann
- Department of Biochemistry & Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA .,Graduate Programs in Genes & Development, University of Texas Graduate School in Biomedical Sciences at Houston, Houston, TX, USA.,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
13
|
A G-Rich Motif in the lncRNA Braveheart Interacts with a Zinc-Finger Transcription Factor to Specify the Cardiovascular Lineage. Mol Cell 2016; 64:37-50. [PMID: 27618485 DOI: 10.1016/j.molcel.2016.08.010] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/21/2016] [Accepted: 08/05/2016] [Indexed: 02/07/2023]
Abstract
Long non-coding RNAs (lncRNAs) are an emerging class of transcripts that can modulate gene expression; however, their mechanisms of action remain poorly understood. Here, we experimentally determine the secondary structure of Braveheart (Bvht) using chemical probing methods and show that this ∼590 nt transcript has a modular fold. Using CRISPR/Cas9-mediated editing of mouse embryonic stem cells, we find that deletion of 11 nt in a 5' asymmetric G-rich internal loop (AGIL) of Bvht (bvhtdAGIL) dramatically impairs cardiomyocyte differentiation. We demonstrate a specific interaction between AGIL and cellular nucleic acid binding protein (CNBP/ZNF9), a zinc-finger protein known to bind single-stranded G-rich sequences. We further show that CNBP deletion partially rescues the bvhtdAGIL mutant phenotype by restoring differentiation capacity. Together, our work shows that Bvht functions with CNBP through a well-defined RNA motif to regulate cardiovascular lineage commitment, opening the door for exploring broader roles of RNA structure in development and disease.
Collapse
|
14
|
Urbanek MO, Jazurek M, Switonski PM, Figura G, Krzyzosiak WJ. Nuclear speckles are detention centers for transcripts containing expanded CAG repeats. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1513-20. [PMID: 27239700 DOI: 10.1016/j.bbadis.2016.05.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/18/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022]
Abstract
The human genetic disorders caused by CAG repeat expansions in the translated sequences of various genes are called polyglutamine (polyQ) diseases because of the cellular "toxicity" of the mutant proteins. The contribution of mutant transcripts to the pathogenesis of these diseases is supported by several observations obtained from cellular models of these disorders. Here, we show that the common feature of cell lines modeling polyQ diseases is the formation of nuclear CAG RNA foci. We performed qualitative and quantitative analyses of these foci in numerous cellular models endogenously and exogenously expressing mutant transcripts by fluorescence in situ hybridization (FISH). We compared the CAG RNA foci of polyQ diseases with the CUG foci of myotonic dystrophy type 1 and found substantial differences in their number and morphology. Smaller differences within the polyQ disease group were also revealed and included a positive correlation between the foci number and the CAG repeat length. We show that expanded CAA repeats, also encoding glutamine, did not trigger RNA foci formation and foci formation is independent of the presence of mutant polyglutamine protein. Using FISH combined with immunofluorescence, we demonstrated partial co-localization of CAG repeat foci with MBNL1 alternative splicing factor, which explains the mild deregulation of MBNL1-dependent genes. We also showed that foci reside within nuclear speckles in diverse cell types: fibroblasts, lymphoblasts, iPS cells and neuronal progenitors and remain dependent on integrity of these nuclear structures.
Collapse
Affiliation(s)
- Martyna O Urbanek
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland
| | - Magdalena Jazurek
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland
| | - Pawel M Switonski
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland
| | - Grzegorz Figura
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland.
| |
Collapse
|
15
|
Urbanek MO, Krzyzosiak WJ. RNA FISH for detecting expanded repeats in human diseases. Methods 2015; 98:115-123. [PMID: 26615955 DOI: 10.1016/j.ymeth.2015.11.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/18/2015] [Accepted: 11/21/2015] [Indexed: 12/14/2022] Open
Abstract
RNA fluorescence in situ hybridization (FISH) is a widely used technique for detecting transcripts in fixed cells and tissues. Many variants of RNA FISH have been proposed to increase signal strength, resolution and target specificity. The current variants of this technique facilitate the detection of the subcellular localization of transcripts at a single molecule level. Among the applications of RNA FISH are studies on nuclear RNA foci in diseases resulting from the expansion of tri-, tetra-, penta- and hexanucleotide repeats present in different single genes. The partial or complete retention of mutant transcripts forming RNA aggregates within the nucleoplasm has been shown in multiple cellular disease models and in the tissues of patients affected with these atypical mutations. Relevant diseases include, among others, myotonic dystrophy type 1 (DM1) with CUG repeats, Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3) with CAG repeats, fragile X-associated tremor/ataxia syndrome (FXTAS) with CGG repeats, myotonic dystrophy type 2 (DM2) with CCUG repeats, amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) with GGGGCC repeats and spinocerebellar ataxia type 32 (SCA32) with GGCCUG. In this article, we summarize the results obtained with FISH to examine RNA nuclear inclusions. We provide a detailed protocol for detecting RNAs containing expanded CAG and CUG repeats in different cellular models, including fibroblasts, lymphoblasts, induced pluripotent stem cells and murine and human neuronal progenitors. We also present the results of the first single-molecule FISH application in a cellular model of polyglutamine disease.
Collapse
Affiliation(s)
- Martyna O Urbanek
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14 Str., 61-704 Poznan, Poland.
| |
Collapse
|
16
|
Holler CJ, Davis PR, Beckett TL, Platt TL, Webb RL, Head E, Murphy MP. Bridging integrator 1 (BIN1) protein expression increases in the Alzheimer's disease brain and correlates with neurofibrillary tangle pathology. J Alzheimers Dis 2015; 42:1221-7. [PMID: 25024306 DOI: 10.3233/jad-132450] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Recent genome wide association studies have implicated bridging integrator 1 (BIN1) as a late-onset Alzheimer's disease (AD) susceptibility gene. There are at least 15 different known isoforms of BIN1, with many being expressed in the brain including the longest isoform (iso1), which is brain-specific and localizes to axon initial segments and nodes of Ranvier. It is currently unknown what role BIN1 plays in AD. We analyzed BIN1 protein expression from a large number (n = 71) of AD cases and controls from five different brain regions (hippocampus, inferior parietal cortex, inferior temporal cortex, frontal cortex (BA9), and superior and middle temporal gyri). We found that the amount of the largest isoform of BIN1 was significantly reduced in the AD brain compared to age-matched controls, and smaller BIN1 isoforms were significantly increased. Further, BIN1 was significantly correlated with the amount of neurofibrillary tangle (NFT) pathology but not with either diffuse or neuritic plaques, or with the amount of amyloid-β peptide. BIN1 is known to be abnormally expressed in another human disease, myotonic dystrophy, which also features prominent NFT pathology. These data suggest that BIN1 is likely involved in AD as a modulator of NFT pathology, and that this role may extend to other human diseases that feature tau pathology.
Collapse
Affiliation(s)
- Christopher J Holler
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Paulina R Davis
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Tina L Beckett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Thomas L Platt
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Robin L Webb
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - M Paul Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA University of Kentucky Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Braun S. Gene-based therapies of neuromuscular disorders: an update and the pivotal role of patient organizations in their discovery and implementation. J Gene Med 2014; 15:397-413. [PMID: 24123726 DOI: 10.1002/jgm.2747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 12/13/2022] Open
Abstract
This review updates the state-of-the art accomplishments of the multifaceted gene-based therapies, which include DNA or RNA as either therapeutic tools or targets for the treatment of neuromuscular diseases. It also provides insights into the key role that patient organizations have played in research and development; in particular, by addressing bottlenecks and generating boundary conditions that have contributed to scientific breakthroughs, and the effectiveness of innovation processes. Several gene therapy methods have reached the clinical stage and are now addressing both specific and classical issues related to this novel technology. Not ready yet for clinical application, genome editing is at its infancy. More rapidly progressing, RNA-based therapeutics, and especially exon skipping, exon inclusion and stop codon readthrough strategies, are about to move to the market. Most importantly, patients were at the forefront of this discovery process, from basic knowledge to innovation and translational research in a rapidly growing field of unmet medical needs. In recent years, Duchenne muscular dystrophy was the fertile ground for new therapeutic concepts that have been extended to other neuromuscular disorders, such as spinal muscular atrophy, myotonic dystrophies or fascioscapulohumeral dystrophy. In line with their longstanding policy, patient organizations will keep working in a proactive manner to bring together all stakeholders with a view to working out truly therapeutic solutions over a long-term perspective.
Collapse
|
20
|
Pettersson OJ, Aagaard L, Andrejeva D, Thomsen R, Jensen TG, Damgaard CK. DDX6 regulates sequestered nuclear CUG-expanded DMPK-mRNA in dystrophia myotonica type 1. Nucleic Acids Res 2014; 42:7186-200. [PMID: 24792155 PMCID: PMC4066779 DOI: 10.1093/nar/gku352] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is caused by CUG triplet expansions in the 3′ UTR of dystrophia myotonica protein kinase (DMPK) messenger ribonucleic acid (mRNA). The etiology of this multi-systemic disease involves pre-mRNA splicing defects elicited by the ability of the CUG-expanded mRNA to ‘sponge’ splicing factors of the muscleblind family. Although nuclear aggregation of CUG-containing mRNPs in distinct foci is a hallmark of DM1, the mechanisms of their homeostasis have not been completely elucidated. Here we show that a DEAD-box helicase, DDX6, interacts with CUG triplet-repeat mRNA in primary fibroblasts from DM1 patients and with CUG–RNA in vitro. DDX6 overexpression relieves DM1 mis-splicing, and causes a significant reduction in nuclear DMPK-mRNA foci. Conversely, knockdown of endogenous DDX6 leads to a significant increase in DMPK-mRNA foci count and to increased sequestration of MBNL1 in the nucleus. While the level of CUG-expanded mRNA is unaffected by increased DDX6 expression, the mRNA re-localizes to the cytoplasm and its interaction partner MBNL1 becomes dispersed and also partially re-localized to the cytoplasm. Finally, we show that DDX6 unwinds CUG-repeat duplexes in vitro in an adenosinetriphosphate-dependent manner, suggesting that DDX6 can remodel and release nuclear DMPK messenger ribonucleoprotein foci, leading to normalization of pathogenic alternative splicing events.
Collapse
Affiliation(s)
- Olof J 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
| | - Diana Andrejeva
- Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, building 1131, DK-8000 Aarhus C, Denmark
| | - Rune Thomsen
- Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, building 1131, DK-8000 Aarhus C, Denmark
| | - Thomas G Jensen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 4, Building 1240, DK-8000 Aarhus C, Denmark
| | - Christian K Damgaard
- Department of Molecular Biology and Genetics, Aarhus University, C.F. Møllers Allé 3, building 1131, DK-8000 Aarhus C, Denmark
| |
Collapse
|
21
|
Jahromi AH, Fu Y, Miller KA, Nguyen L, Luu LM, Baranger AM, Zimmerman SC. Developing bivalent ligands to target CUG triplet repeats, the causative agent of myotonic dystrophy type 1. J Med Chem 2013; 56:9471-9481. [PMID: 24188018 DOI: 10.1021/jm400794z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An expanded CUG repeat transcript (CUG(exp)) is the causative agent of myotonic dystrophy type 1 (DM1) by sequestering muscleblind-like 1 protein (MBNL1), a regulator of alternative splicing. On the basis of a ligand (1) that was previously reported to be active in an in vitro assay, we present the synthesis of a small library containing 10 dimeric ligands (4-13) that differ in length, composition, and attachment point of the linking chain. The oligoamino linkers gave a greater gain in affinity for CUG RNA and were more effective when compared to oligoether linkers. The most potent in vitro ligand (9) was shown to be aqueous-soluble and both cell- and nucleus-permeable, displaying almost complete dispersion of MBNL1 ribonuclear foci in a DM1 cell model. Direct evidence for the bioactivity of 9 was observed in its ability to disperse ribonuclear foci in individual live DM1 model cells using time-lapse confocal fluorescence microscopy.
Collapse
Affiliation(s)
- Amin Haghighat Jahromi
- Center for Biophysics and Computational Biology, University of Illinois, Urbana, IL, USA.,Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Yuan Fu
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Kali A Miller
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Lien Nguyen
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Long M Luu
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Anne M Baranger
- Department of Chemistry, University of Illinois, Urbana, IL, USA
| | - Steven C Zimmerman
- Center for Biophysics and Computational Biology, University of Illinois, Urbana, IL, USA.,Department of Chemistry, University of Illinois, Urbana, IL, USA
| |
Collapse
|
22
|
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
|
23
|
Sicot G, Gomes-Pereira M. RNA toxicity in human disease and animal models: from the uncovering of a new mechanism to the development of promising therapies. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1390-409. [PMID: 23500957 DOI: 10.1016/j.bbadis.2013.03.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 03/01/2013] [Accepted: 03/04/2013] [Indexed: 01/06/2023]
Abstract
Mutant ribonucleic acid (RNA) molecules can be toxic to the cell, causing human disease through trans-acting dominant mechanisms. RNA toxicity was first described in myotonic dystrophy type 1, a multisystemic disorder caused by the abnormal expansion of a non-coding trinucleotide repeat sequence. The development of multiple and complementary animal models of disease has greatly contributed to clarifying the complex disease pathways mediated by toxic RNA molecules. RNA toxicity is not limited to myotonic dystrophy and spreads to an increasing number of human conditions, which share some unifying pathogenic events mediated by toxic RNA accumulation and disruption of RNA-binding proteins. The remarkable progress in the dissection of disease pathobiology resulted in the rational design of molecular therapies, which have been successfully tested in animal models. Toxic RNA diseases, and in particular myotonic dystrophy, clearly illustrate the critical contribution of animal models of disease in translational research: from gene mutation to disease mechanisms, and ultimately to therapy development. This article is part of a Special Issue entitled: Animal Models of Disease.
Collapse
|
24
|
Gao Z, Cooper TA. Antisense oligonucleotides: rising stars in eliminating RNA toxicity in myotonic dystrophy. Hum Gene Ther 2013; 24:499-507. [PMID: 23252746 DOI: 10.1089/hum.2012.212] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myotonic dystrophy (DM) is a dominantly inherited, multisystemic disease caused by expanded CTG (type 1, DM1) or CCTG (type 2, DM2) repeats in untranslated regions of the mutated genes. Pathogenesis results from expression of RNAs from the mutated alleles that are toxic because of the expanded CUG or CCUG repeats. Increased understanding of the repeat-containing RNA (C/CUG(exp) RNA)-induced toxicity has led to the development of multiple strategies targeting the toxic RNA. Among these approaches, antisense oligonucleotides (ASOs) have demonstrated high potency in reversing the RNA toxicity in both cultured DM1 cells and DM1 animal models, thus offering great promise for the potential treatment of DM1. ASO targeting approaches will also provide avenues for the treatment of other repeat RNA-mediated diseases.
Collapse
Affiliation(s)
- Zhihua Gao
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
25
|
Jones K, Wei C, Iakova P, Bugiardini E, Schneider-Gold C, Meola G, Woodgett J, Killian J, Timchenko NA, Timchenko LT. GSK3β mediates muscle pathology in myotonic dystrophy. J Clin Invest 2012; 122:4461-72. [PMID: 23160194 DOI: 10.1172/jci64081] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 09/21/2012] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a complex neuromuscular disease characterized by skeletal muscle wasting, weakness, and myotonia. DM1 is caused by the accumulation of CUG repeats, which alter the biological activities of RNA-binding proteins, including CUG-binding protein 1 (CUGBP1). CUGBP1 is an important skeletal muscle translational regulator that is activated by cyclin D3-dependent kinase 4 (CDK4). Here we show that mutant CUG repeats suppress Cdk4 signaling by increasing the stability and activity of glycogen synthase kinase 3β (GSK3β). Using a mouse model of DM1 (HSA(LR)), we found that CUG repeats in the 3' untranslated region (UTR) of human skeletal actin increase active GSK3β in skeletal muscle of mice, prior to the development of skeletal muscle weakness. Inhibition of GSK3β in both DM1 cell culture and mouse models corrected cyclin D3 levels and reduced muscle weakness and myotonia in DM1 mice. Our data predict that compounds normalizing GSK3β activity might be beneficial for improvement of muscle function in patients with DM1.
Collapse
Affiliation(s)
- Karlie Jones
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Udd B, Krahe R. The myotonic dystrophies: molecular, clinical, and therapeutic challenges. Lancet Neurol 2012; 11:891-905. [DOI: 10.1016/s1474-4422(12)70204-1] [Citation(s) in RCA: 334] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
The impact of mRNA turnover and translation on age-related muscle loss. Ageing Res Rev 2012; 11:432-41. [PMID: 22687959 DOI: 10.1016/j.arr.2012.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/25/2012] [Accepted: 05/31/2012] [Indexed: 12/21/2022]
Abstract
The deterioration of skeletal muscle that develops slowly with age, termed sarcopenia, often leads to disability and mortality in the elderly population. As the proportion of elderly citizens continues to increase due to the dramatic rise in life expectancy, there are rising concerns about the healthcare cost and social burden of caring for geriatric patients. Thus, there is a growing need to understand the underlying mechanisms of sarcopenic muscle loss so that more efficacious therapies may be developed. Building evidence suggests that the onset of age-related muscle loss is linked to the age-related changes in gene expression that occur during sarcopenia. In recent work, the posttranscriptional regulation of gene expression by RNA-binding proteins (RBPs) and microRNA (miRNA) involved in the turnover and translation of mRNA were shown as key players believed to be involved in the induction of muscle wasting. Furthermore, posttranscriptional regulation may also be linked to the reduced ability of muscle satellite cells to contribute to muscle mass during ageing, a key contributing factor to sarcopenic progression. Here we highlight how the activation of pathways such as the p38 MAPK and the phosphoinositide 3-kinase (PI3K) pathways alter the ability of RBPs to regulate the expression of their target mRNAs encoding proteins involved in cell cycle (p21 and p16), as well as myogenesis (Pax7, myogenin and MyoD). Further investigation into the role of RBPs and miRNA during sarcopenia may provide new insights into the development and progression of this disorder, which may lead to the development of new treatment options for elderly patients suffering from sarcopenia.
Collapse
|
28
|
Jones K, Timchenko L, Timchenko NA. The role of CUGBP1 in age-dependent changes of liver functions. Ageing Res Rev 2012; 11:442-9. [PMID: 22446383 DOI: 10.1016/j.arr.2012.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 02/09/2012] [Accepted: 02/10/2012] [Indexed: 12/14/2022]
Abstract
Aging liver is characterized by alterations of liver biology and by a reduction of many functions which are important for the maintenance of body homeostasis. The main dysfunctions include appearance of enlarged hepatocytes, impaired liver regeneration after partial hepatectomy (PH), development of hepatic steatosis, reduction of secretion of proteins and alterations in the hepatic sinusoid. RNA binding proteins are involved in the regulation of gene expression in all tissues including regulation of biological processes in the liver. This review is focused on the role of a conserved, multi-functional RNA-binding protein, CUGBP1, in the development of aging phenotype in the liver. CUGBP1 has been identified as a protein which binds to RNA CUG repeats expanded in Myotonic Dystrophy type 1 (DM1). CUGBP1 is highly expressed in the liver and regulates translation of proteins which are critical for maintenance of liver functions. In livers of young mice, CUGBP1 forms complexes with eukaryotic translation initiation factor eIF2 and supports translation of C/EBPβ and HDAC1 proteins, which are involved in liver growth, differentiation and liver cancer. Aging changes several signaling pathways which lead to the elevation of the CUGBP1-eIF2α complex and to an increase of translation of C/EBPβ and HDAC1. These proteins form multi-protein complexes with additional transcription factors and with chromatin remodeling proteins causing epigenetic alterations of gene expression in livers of old mice. It appears that CUGBP1-mediated translational elevation of HDAC1 is one of the key events in the epigenetic changes in livers of old mice, leading to the development of age-associated dysfunctions of the liver. This review will also discuss a possible role of CUGBP1 in liver dysfunction in patients affected with DM1.
Collapse
|
29
|
Malatesta M. Skeletal muscle features in myotonic dystrophy and sarcopenia: do similar nuclear mechanisms lead to skeletal muscle wasting? Eur J Histochem 2012; 56:e36. [PMID: 23027352 PMCID: PMC3493982 DOI: 10.4081/ejh.2012.e36] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 08/03/2012] [Indexed: 02/06/2023] Open
Abstract
In the cell nucleus, the gene primary transcripts undergo molecular processing to generate mature RNAs, which are finally exported to the cytoplasm. These mRNA maturation events are chronologically and spatially ordered, and mostly occur on distinct ribonucleoprotein (RNP)-containing structures. Defects in the mRNA maturation pathways have been demonstrated in myotonic dystrophy type 1 (DM1) and type 2 (DM2) whose characteristic multisystemic features are caused by the expansion of two distinct nucleotide sequences: (CTG)n in the DMPK gene on chromosome 19q13 in DM1, and (CCTG)n in the ZNF9 gene on chromosome 3q21 in DM2. By combining biomolecular and cytochemical techniques, it has been shown that the basic mechanisms of DMs reside in the accumulation of CUG- or CCUG-containing transcripts in intranuclear foci where several RNA-binding proteins necessary for the physiological processing of pre-mRNA are sequestered. Moreover, a nucleoplasmic accumulation of splicing and cleavage factors has been found in DMs. This suggests that the dystrophic phenotype could depend on a general alteration of the pre-mRNA post-transcriptional pathway. Interestingly, the accumulation of pre-mRNA processing factors in the myonuclei of DM1 and DM2 patients is reminiscent of the nuclear alterations typical of sarcopenia, i.e., the loss of muscle mass and function which physiologically occurs during ageing. Consistently, in an in vitro study, we observed that satellite-cell-derived DM2 myoblasts show cell senescence alterations and impairment of the pre-mRNA maturation pathways earlier than the myoblasts from healthy patient. These results suggest possible common cellular mechanisms responsible for skeletal muscle wasting in sarcopenia and in myotonic dystrophy.
Collapse
Affiliation(s)
- M Malatesta
- Dipartimento di Scienze Neurologiche, Neuropsicologiche, Morfologiche e Motorie, Sezione di Anatomia e Istologia, Università degli Studi di Verona, Strada Le Grazie 8, Verona, Italy.
| |
Collapse
|
30
|
Ravel-Chapuis A, Bélanger G, Yadava RS, Mahadevan MS, DesGroseillers L, Côté J, Jasmin BJ. The RNA-binding protein Staufen1 is increased in DM1 skeletal muscle and promotes alternative pre-mRNA splicing. ACTA ACUST UNITED AC 2012; 196:699-712. [PMID: 22431750 PMCID: PMC3308689 DOI: 10.1083/jcb.201108113] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Staufen1 interacts with mRNAs with expanded CUG repeats and promotes their nuclear export and translation, while also promoting alternative splicing of other mRNAs. In myotonic dystrophy type 1 (DM1), dystrophia myotonica protein kinase messenger ribonucleic acids (RNAs; mRNAs) with expanded CUG repeats (CUGexp) aggregate in the nucleus and become toxic to cells by sequestering and/or misregulating RNA-binding proteins, resulting in aberrant alternative splicing. In this paper, we find that the RNA-binding protein Staufen1 is markedly and specifically increased in skeletal muscle from DM1 mouse models and patients. We show that Staufen1 interacts with mutant CUGexp mRNAs and promotes their nuclear export and translation. This effect is critically dependent on the third double-stranded RNA–binding domain of Staufen1 and shuttling of Staufen1 into the nucleus via its nuclear localization signal. Moreover, we uncover a new role of Staufen1 in splicing regulation. Overexpression of Staufen1 rescues alternative splicing of two key pre-mRNAs known to be aberrantly spliced in DM1, suggesting its increased expression represents an adaptive response to the pathology. Altogether, our results unravel a novel function for Staufen1 in splicing regulation and indicate that it may positively modulate the complex DM1 phenotype, thereby revealing its potential as a therapeutic target.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | | | | | | | | | | | | |
Collapse
|
31
|
Nuclear ribonucleoprotein-containing foci increase in size in non-dividing cells from patients with myotonic dystrophy type 2. Histochem Cell Biol 2012; 138:699-707. [PMID: 22706481 DOI: 10.1007/s00418-012-0984-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2012] [Indexed: 10/28/2022]
Abstract
Myotonic dystrophies (DM) are genetically based neuromuscular disorders characterized by the accumulation of mutant transcripts into peculiar intranuclear foci, where different splicing factors (among which the alternative splicing regulator muscleblind-like 1 protein, MBNL1) are ectopically sequestered. The aim of the present investigation was to describe the dynamics of the DM-specific intranuclear foci in interphase nuclei and during mitosis, as well as after the exit from the cell cycle. Primary cultures of skin fibroblasts from DM2 patients were used, as a model system to reproduce in vitro, as accurately as possible, the in vivo conditions. Cycling and resting fibroblasts were investigated by immunocytochemical and morphometric techniques, and the relative amounts of MBNL1 were also estimated by western blotting. MBNL1-containing foci were exclusively found in the nucleus during most of the interphase, while being observed in the cytoplasm during mitosis when they never associate with the chromosomes; the foci remained in the cytoplasm at cytodieresis, and underwent disassembly in early G1 to be reformed in the nucleus at each cell cycle. After fibroblasts had stopped dividing in late-passage cultures, the nuclear foci were observed to progressively increase in size. Interestingly, measurements on muscle biopsies taken from the same DM2 patients at different ages demonstrated that, in the nuclei of myofibers, the MBNL1-containing foci become larger with increasing patient's age. As a whole, these results suggest that in non-dividing cells of DM2 patients the sequestration in the nuclear foci of factors needed for RNA processing would be continuous and progressive, eventually leading to the onset (and the worsening with time) of the pathological traits. This is consistent with the evidence that in DM patients the most affected organs or tissues are those where non-renewing cells are mainly present, i.e., the central nervous system, heart and skeletal muscle.
Collapse
|