49
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Molecular, Physiological, and Motor Performance Defects in DMSXL Mice Carrying >1,000 CTG Repeats from the Human DM1 Locus

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Myotonic dystrophy type 1 (DM1) is caused by an unstable CTG repeat expansion in the 3′UTR of the DM protein kinase ( DMPK) gene. DMPK transcripts carrying CUG expansions form nuclear foci and affect splicing regulation of various RNA transcripts. Furthermore, bidirectional transcription over the DMPK gene and non-conventional RNA translation of repeated transcripts have been described in DM1. It is clear now that this disease may involve multiple pathogenic pathways including changes in gene expression, RNA stability and splicing regulation, protein translation, and micro–RNA metabolism. We previously generated transgenic mice with 45-kb of the DM1 locus and >300 CTG repeats (DM300 mice). After successive breeding and a high level of CTG repeat instability, we obtained transgenic mice carrying >1,000 CTG (DMSXL mice). Here we described for the first time the expression pattern of the DMPK sense transcripts in DMSXL and human tissues. Interestingly, we also demonstrate that DMPK antisense transcripts are expressed in various DMSXL and human tissues, and that both sense and antisense transcripts accumulate in independent nuclear foci that do not co-localize together. Molecular features of DM1-associated RNA toxicity in DMSXL mice (such as foci accumulation and mild missplicing), were associated with high mortality, growth retardation, and muscle defects (abnormal histopathology, reduced muscle strength, and lower motor performances). We have found that lower levels of IGFBP-3 may contribute to DMSXL growth retardation, while increased proteasome activity may affect muscle function. These data demonstrate that the human DM1 locus carrying very large expansions induced a variety of molecular and physiological defects in transgenic mice, reflecting DM1 to a certain extent. As a result, DMSXL mice provide an animal tool to decipher various aspects of the disease mechanisms. In addition, these mice can be used to test the preclinical impact of systemic therapeutic strategies on molecular and physiological phenotypes.

          Author Summary

          Myotonic dystrophy type 1 (DM1) is caused by the abnormal expansion of a CTG repeat located in the DM protein kinase ( DMPK) gene. DMPK transcripts carrying CUG expansions form toxic nuclear foci that affect other RNAs. DM1 involve multiple pathogenic pathways including changes in gene expression, RNA stability and splicing regulation, protein translation, and micro–RNA metabolism. We previously generated transgenic mice carrying the human DM1 locus and very large expansions >1,000 CTG (DMSXL mice). Here we described for the first time, the expression pattern of the DMPK sense transcripts in DMSXL and human tissues. We also demonstrate that DMPK antisense transcripts are expressed in various tissues from DMSXL mice and human. Both sense and antisense transcripts form nuclear foci. DMSXL mice showed molecular DM1 features such as foci and mild splicing defects as well as muscles defects, reduced muscle strength, and lower motor performances. These mice recapitulate some molecular features of DM1 leading to physiological abnormalities. DMSXL are not only a tool to decipher various mechanisms involved in DM1 but also to test the preclinical impact of systemic therapeutic strategies.

          Related collections

          Most cited references42

          • Record: found
          • Abstract: found
          • Article: not found

          Molecular basis of myotonic dystrophy: expansion of a trinucleotide (CTG) repeat at the 3' end of a transcript encoding a protein kinase family member.

          Using positional cloning strategies, we have identified a CTG triplet repeat that undergoes expansion in myotonic dystrophy patients. This sequence is highly variable in the normal population. PCR analysis of the interval containing this repeat indicates that unaffected individuals have been 5 and 27 copies. Myotonic dystrophy patients who are minimally affected have at least 50 repeats, while more severely affected patients have expansion of the repeat containing segment up to several kilobase pairs. The CTG repeat is transcribed and is located in the 3' untranslated region of an mRNA that is expressed in tissues affected by myotonic dystrophy. This mRNA encodes a polypeptide that is a member of the protein kinase family.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Recruitment of human muscleblind proteins to (CUG)(n) expansions associated with myotonic dystrophy.

            Myotonic dystrophy (DM1) is an autosomal dominant neuromuscular disorder associated with a (CTG)(n) expansion in the 3'-untranslated region of the DM1 protein kinase (DMPK) gene. To explain disease pathogenesis, the RNA dominance model proposes that the DM1 mutation produces a gain-of-function at the RNA level in which CUG repeats form RNA hairpins that sequester nuclear factors required for proper muscle development and maintenance. Here, we identify the triplet repeat expansion (EXP) RNA-binding proteins as candidate sequestered factors. As predicted by the RNA dominance model, binding of the EXP proteins is specific for dsCUG RNAs and proportional to the size of the triplet repeat expansion. Remarkably, the EXP proteins are homologous to the Drosophila muscleblind proteins required for terminal differentiation of muscle and photoreceptor cells. EXP expression is also activated during mammalian myoblast differentiation, but the EXP proteins accumulate in nuclear foci in DM1 cells. We propose that DM1 disease is caused by aberrant recruitment of the EXP proteins to the DMPK transcript (CUG)(n) expansion.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Muscleblind-like 2-mediated alternative splicing in the developing brain and dysregulation in myotonic dystrophy.

              The RNA-mediated disease model for myotonic dystrophy (DM) proposes that microsatellite C(C)TG expansions express toxic RNAs that disrupt splicing regulation by altering MBNL1 and CELF1 activities. While this model explains DM manifestations in muscle, less is known about the effects of C(C)UG expression on the brain. Here, we report that Mbnl2 knockout mice develop several DM-associated central nervous system (CNS) features including abnormal REM sleep propensity and deficits in spatial memory. Mbnl2 is prominently expressed in the hippocampus and Mbnl2 knockouts show a decrease in NMDA receptor (NMDAR) synaptic transmission and impaired hippocampal synaptic plasticity. While Mbnl2 loss did not significantly alter target transcript levels in the hippocampus, misregulated splicing of hundreds of exons was detected using splicing microarrays, RNA-seq, and HITS-CLIP. Importantly, the majority of the Mbnl2-regulated exons examined were similarly misregulated in DM. We propose that major pathological features of the DM brain result from disruption of the MBNL2-mediated developmental splicing program. Copyright © 2012 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                November 2012
                November 2012
                29 November 2012
                : 8
                : 11
                : e1003043
                Affiliations
                [1 ]Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
                [2 ]Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
                [3 ]Généthon, Evry, France
                [4 ]Inserm U955, Département de Neurosciences, Faculté de Médecine, Université Paris XII, Créteil, France
                [5 ]Université Paris Descartes-Sorbonne Paris Cité, Paris, France
                [6 ]F. Hoffmann-La Roche, CNS Pharma Research and Development, Basel, Switzerland
                [7 ]Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
                [8 ]UPMC Univ Paris 06, UM 76, Institut de Myologie and Inserm, U974 and CNRS, UMR7215, Paris, France
                [9 ]Human Genetics Research Unit, Laval University, Québec City, Québec, Canada
                The Hospital for Sick Children and University of Toronto, Canada
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: A Ferry, J-Y Hogrel, F Metzger, J Puymirat, G Bassez, D Furling, G Gourdon. Performed the experiments: A Huguet, F Medja, A Nicole, A Vignaud, C Guiraud-Dogan, A Ferry, V Decostre, J-Y Hogrel, F Metzger, A Hoeflich, M Baraibar, M Gomes-Pereira. Analyzed the data: A Huguet, F Medja, C Guiraud-Dogan, A Ferry, V Decostre, J-Y Hogrel, F Metzger, A Hoeflich, G Bassez, D Furling, G Gourdon. Contributed reagents/materials/analysis tools: F Metzger, A Hoeflich, M Baraibar, G Bassez, D Furling, A Munnich, G Gourdon. Wrote the paper: A Huguet, F Medja, V Decostre, J-Y Hogrel, F Metzger, A Hoeflich, M Gomes-Pereira, D Furling, G Gourdon.

                Article
                PGENETICS-D-12-00540
                10.1371/journal.pgen.1003043
                3510028
                23209425
                1c92bbc6-4cc0-45be-8a31-36fe8c6c883d
                Copyright @ 2012

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 2 March 2012
                : 5 September 2012
                Page count
                Pages: 19
                Funding
                This work was supported by ANR (Agence Nationale de Recherche, France; DM1MICE project), AFM (Association Française contre les Myopathies, France), Inserm (Institute National de la Santé et Recherche Médicale, France), and Université Paris Descartes (Paris, France). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology
                Genetics
                Model Organisms
                Molecular Cell Biology

                Genetics
                Genetics

                Comments

                Comment on this article