51
|
Matloka M, Klein AF, Rau F, Furling D. Cells of Matter- In Vitro Models for Myotonic Dystrophy. Front Neurol 2018; 9:361. [PMID: 29875732 PMCID: PMC5974047 DOI: 10.3389/fneur.2018.00361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1 also known as Steinert disease) is a multisystemic disorder mainly characterized by myotonia, progressive muscle weakness and wasting, cognitive impairments, and cardiac defects. This autosomal dominant disease is caused by the expression of nuclear retained RNAs containing pathologic expanded CUG repeats that alter the function of RNA-binding proteins in a tissue-specific manner, leading ultimately to neuromuscular dysfunction and clinical symptoms. Although considerable knowledge has been gathered on myotonic dystrophy since its first description, the development of novel relevant disease models remains of high importance to investigate pathophysiologic mechanisms and to assess new therapeutic approaches. In addition to animal models, in vitro cell cultures provide a unique resource for both fundamental and translational research. This review discusses how cellular models broke ground to decipher molecular basis of DM1 and describes currently available cell models, ranging from exogenous expression of the CTG tracts to variable patients' derived cells.
Collapse
Affiliation(s)
| | | | | | - Denis Furling
- Sorbonne Université, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
52
|
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
|
53
|
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
|
54
|
Traverso M, Assereto S, Baratto S, Iacomino M, Pedemonte M, Diana MC, Ferretti M, Broda P, Minetti C, Gazzerro E, Madia F, Bruno C, Zara F, Fiorillo C. Clinical and molecular consequences of exon 78 deletion in DMD gene. J Hum Genet 2018; 63:761-764. [PMID: 29556034 DOI: 10.1038/s10038-018-0439-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 01/19/2023]
Abstract
We present a 13-year-old patient with persistent increase of serum Creatine Kinase (CK) and myalgia after exertion. Skeletal muscle biopsy showed marked reduction of dystrophin expression leading to genetic analysis of DMD gene by MLPA, which detected a single deletion of exon 78. To the best of our knowledge, DMD exon 78 deletion has never been described in literature and, according to prediction, it should lead to loss of reading frame in the dystrophin gene. To further assess the actual effect of exon 78 deletion, we analysed cDNA from muscle mRNA. This analysis confirmed the absence of 32 bp of exon 78. Exclusion of exon 78 changes the open reading frame of exon 79 and generate a downstream stop codon, producing a dystrophin protein of 3703 amino acids instead of 3685 amino acids. Albeit loss of reading frame usually leads to protein degradation and severe phenotype, in this case, we demonstrated that deletion of DMD exon 78 can be associated with a functional protein able to bind DGC complex and a very mild phenotype. This study adds a novel deletion in DMD gene in human and helps to define the compliance between maintaining/disrupting the reading frame and clinical form of the disease.
Collapse
Affiliation(s)
| | - Stefania Assereto
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Serena Baratto
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | | | - Marina Pedemonte
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Maria Cristina Diana
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Marta Ferretti
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Paolo Broda
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Carlo Minetti
- DINOGMI, University of Genoa, Genoa, Italy.,Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Elisabetta Gazzerro
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Francesca Madia
- Laboratory of Neurogenetic and Neuroscience, G. Gaslini Institute, Genoa, Italy
| | - Claudio Bruno
- Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy
| | - Federico Zara
- Laboratory of Neurogenetic and Neuroscience, G. Gaslini Institute, Genoa, Italy
| | - Chiara Fiorillo
- DINOGMI, University of Genoa, Genoa, Italy. .,Paediatric Neurology and Muscular Diseases Unit, University of Genoa and G. Gaslini Institute, Genoa, Italy.
| |
Collapse
|
55
|
Nakka K, Ghigna C, Gabellini D, Dilworth FJ. Diversification of the muscle proteome through alternative splicing. Skelet Muscle 2018; 8:8. [PMID: 29510724 PMCID: PMC5840707 DOI: 10.1186/s13395-018-0152-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/15/2018] [Indexed: 12/16/2022] Open
Abstract
Background Skeletal muscles express a highly specialized proteome that allows the metabolism of energy sources to mediate myofiber contraction. This muscle-specific proteome is partially derived through the muscle-specific transcription of a subset of genes. Surprisingly, RNA sequencing technologies have also revealed a significant role for muscle-specific alternative splicing in generating protein isoforms that give specialized function to the muscle proteome. Main body In this review, we discuss the current knowledge with respect to the mechanisms that allow pre-mRNA transcripts to undergo muscle-specific alternative splicing while identifying some of the key trans-acting splicing factors essential to the process. The importance of specific splicing events to specialized muscle function is presented along with examples in which dysregulated splicing contributes to myopathies. Though there is now an appreciation that alternative splicing is a major contributor to proteome diversification, the emergence of improved “targeted” proteomic methodologies for detection of specific protein isoforms will soon allow us to better appreciate the extent to which alternative splicing modifies the activity of proteins (and their ability to interact with other proteins) in the skeletal muscle. In addition, we highlight a continued need to better explore the signaling pathways that contribute to the temporal control of trans-acting splicing factor activity to ensure specific protein isoforms are expressed in the proper cellular context. Conclusions An understanding of the signal-dependent and signal-independent events driving muscle-specific alternative splicing has the potential to provide us with novel therapeutic strategies to treat different myopathies. Electronic supplementary material The online version of this article (10.1186/s13395-018-0152-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kiran Nakka
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Claudia Ghigna
- Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), Pavia, Italy
| | - Davide Gabellini
- Unit of Gene Expression and Muscular Dystrophy, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, DIBIT2, 5A3-44, via Olgettina 58, 20132, Milan, Italy.
| | - F Jeffrey Dilworth
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada. .,Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Rd, Mailbox 511, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
56
|
Charton K, Suel L, Henriques SF, Moussu JP, Bovolenta M, Taillepierre M, Becker C, Lipson K, Richard I. Exploiting the CRISPR/Cas9 system to study alternative splicing in vivo: application to titin. Hum Mol Genet 2018; 25:4518-4532. [PMID: 28173117 DOI: 10.1093/hmg/ddw280] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/29/2016] [Accepted: 08/18/2016] [Indexed: 11/12/2022] Open
Abstract
The giant protein titin is the third most abundant protein in striated muscle. Mutations in its gene are responsible for diseases affecting the cardiac and/or the skeletal muscle. Titin has been reported to be expressed in multiple isoforms with considerable variability in the I-band, ensuring the modulation of the passive mechanical properties of the sarcomere. In the M-line, only the penultimate Mex5 exon coding for the specific is7 domain has been reported to be subjected to alternative splicing. Using the CRISPR-Cas9 editing technology, we generated a mouse model where we stably prevent the expression of alternative spliced variant(s) carrying the corresponding domain. Interestingly, the suppression of the domain induces a phenotype mostly in tissues usually expressing the isoform that has been suppressed, indicating that it fulfills (a) specific function(s) in these tissues allowing a perfect adaptation of the M-line to physiological demands of different muscles.
Collapse
Affiliation(s)
- Karine Charton
- INSERM, U951, INTEGRARE research unit Evry, France,Généthon, Evry, France
| | - Laurence Suel
- INSERM, U951, INTEGRARE research unit Evry, France,Généthon, Evry, France
| | - Sara F Henriques
- INSERM, U951, INTEGRARE research unit Evry, France,Généthon, Evry, France,University of Evry-Val-D’Essone, Evry, France
| | - Jean-Paul Moussu
- SEAT - SErvice des Animaux Transgéniques CNRS -TAAM -phenomin UPS44 Bâtiment G 7, rue Guy Môquet 94800 Villejuif, France
| | - Matteo Bovolenta
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Miguel Taillepierre
- SEAT - SErvice des Animaux Transgéniques CNRS -TAAM -phenomin UPS44 Bâtiment G 7, rue Guy Môquet 94800 Villejuif, France
| | - Céline Becker
- SEAT - SErvice des Animaux Transgéniques CNRS -TAAM -phenomin UPS44 Bâtiment G 7, rue Guy Môquet 94800 Villejuif, France
| | - Karelia Lipson
- SEAT - SErvice des Animaux Transgéniques CNRS -TAAM -phenomin UPS44 Bâtiment G 7, rue Guy Môquet 94800 Villejuif, France
| | - Isabelle Richard
- INSERM, U951, INTEGRARE research unit Evry, France,Généthon, Evry, France
| |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW This article describes clinical and electrical myotonia and provides an update on the classification, diagnosis, and management of myotonic disorders. RECENT FINDINGS In the myotonic dystrophies, antisense oligonucleotides provide a general strategy to correct RNA gain of function and modulate the expression of CTG expanded repeats; they are currently being tested in a phase 1-2 randomized controlled trial in patients with adult-onset myotonic dystrophy type 1. New genetic mutations are continuously being identified in the nondystrophic myotonias involving sodium and chloride channels. This contributes to the difficulty in describing genotype-phenotype correlations as the same mutations can give rise to different phenotypes, and the same phenotypes can arise from different mutations. Pharmacologic therapy is moving toward mutation-targeted treatments. SUMMARY This article describes the clinical and diagnostic characteristics and management of the myotonic dystrophies and the nondystrophic myotonias. Clinical features of the congenital, juvenile, and classic adult forms of myotonic dystrophy type 1 are reviewed, and for the adult form, reference is made to the main diagnostic and follow-up tests for which general consensus exists. The different clinical presentations of myotonic dystrophy type 2 and its main differential diagnostic options are also discussed. The clinical spectrum of the sodium and chloride channelopathies is described, and clinical diagnostic clues to differentiate between these two groups are provided. Therapeutic options for patients with nondystrophic myotonias are also presented with reference to literature review and the author's personal experience.
Collapse
|
58
|
An Overview of Recent Advances and Clinical Applications of Exon Skipping and Splice Modulation for Muscular Dystrophy and Various Genetic Diseases. Methods Mol Biol 2018; 1828:31-55. [PMID: 30171533 DOI: 10.1007/978-1-4939-8651-4_2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Exon skipping is a therapeutic approach that is feasible for various genetic diseases and has been studied and developed for over two decades. This approach uses antisense oligonucleotides (AON) to modify the splicing of pre-mRNA to correct the mutation responsible for a disease, or to suppress a particular gene expression, as in allergic diseases. Antisense-mediated exon skipping is most extensively studied in Duchenne muscular dystrophy (DMD) and has developed from in vitro proof-of-concept studies to clinical trials targeting various single exons such as exon 45 (casimersen), exon 53 (NS-065/NCNP-01, golodirsen), and exon 51 (eteplirsen). Eteplirsen (brand name Exondys 51), is the first approved antisense therapy for DMD in the USA, and provides a treatment option for ~14% of all DMD patients, who are amenable to exon 51 skipping. Eteplirsen is granted accelerated approval and marketing authorization by the US Food and Drug Administration (FDA), on the condition that additional postapproval trials show clinical benefit. Permanent exon skipping achieved at the DNA level using clustered regularly interspaced short palindromic repeats (CRISPR) technology holds promise in current preclinical trials for DMD. In hopes of achieving clinical success parallel to DMD, exon skipping and splice modulation are also being studied in other muscular dystrophies, such as Fukuyama congenital muscular dystrophy (FCMD), dysferlinopathy including limb-girdle muscular dystrophy type 2B (LGMD2B), Miyoshi myopathy (MM), and distal anterior compartment myopathy (DMAT), myotonic dystrophy, and merosin-deficient congenital muscular dystrophy type 1A (MDC1A). This chapter also summarizes the development of antisense-mediated exon skipping therapy in diseases such as Usher syndrome, dystrophic epidermolysis bullosa, fibrodysplasia ossificans progressiva (FOP), and allergic diseases.
Collapse
|
59
|
Roos A, Thompson R, Horvath R, Lochmüller H, Sickmann A. Intersection of Proteomics and Genomics to "Solve the Unsolved" in Rare Disorders such as Neurodegenerative and Neuromuscular Diseases. Proteomics Clin Appl 2017; 12. [PMID: 29059504 DOI: 10.1002/prca.201700073] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/30/2017] [Indexed: 01/10/2023]
Abstract
Despite recent rapid advances in sequencing technologies, a significant proportion of patients with rare genetic disorders do not receive a genetic diagnosis after exhaustive testing, and even fewer have a potential therapeutic target identified. Taking rare neuromuscular and neurodegenerative disorders as a paradigm that can be extended to other rare Mendelian disorders, this viewpoint explores the opportunities that are brought about by the integration of genomics and proteomics, as well as the limitations and remaining challenges of this newly emerging field of proteogenomics. The relevance of combining proteomic findings with genetic results for diagnosis and gene discovery is illustrated, highlighting the insights the combined analysis provides into the underlying biology and aetiology as well as the limitations of the experimental techniques. A final discussion focuses on the importance of mechanisms to enable the sharing, reuse, and analysis of source experimental data and describes some of the international initiatives that are making progress in this area.
Collapse
Affiliation(s)
- Andreas Roos
- John Walton Muscular Dystrophy Research Centre, International Centre for Life, Institute of Genetic Medicine, Newcastle upon Tyne, England, UK.,Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Rachel Thompson
- John Walton Muscular Dystrophy Research Centre, International Centre for Life, Institute of Genetic Medicine, Newcastle upon Tyne, England, UK
| | - Rita Horvath
- John Walton Muscular Dystrophy Research Centre, International Centre for Life, Institute of Genetic Medicine, Newcastle upon Tyne, England, UK
| | - Hanns Lochmüller
- John Walton Muscular Dystrophy Research Centre, International Centre for Life, Institute of Genetic Medicine, Newcastle upon Tyne, England, UK
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,Medizinisches Proteom-Center (MPC), Ruhr-Universitat Bochum, Bochum, Germany
| |
Collapse
|
60
|
Konieczny P, Stepniak-Konieczna E, Taylor K, Sznajder LJ, Sobczak K. Autoregulation of MBNL1 function by exon 1 exclusion from MBNL1 transcript. Nucleic Acids Res 2017; 45:1760-1775. [PMID: 27903900 PMCID: PMC5389549 DOI: 10.1093/nar/gkw1158] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 11/07/2016] [Indexed: 01/14/2023] Open
Abstract
Muscleblind-like proteins (MBNLs) are regulators of RNA metabolism. During tissue differentiation the level of MBNLs increases, while their functional insufficiency plays a crucial role in myotonic dystrophy (DM). Deep sequencing of RNA molecules cross-linked to immunoprecipitated protein particles (CLIP-seq) revealed that MBNL1 binds to MBNL1 exon 1 (e1) encoding both the major part of 5΄UTR and an amino-terminal region of MBNL1 protein. We tested several hypotheses regarding the possible autoregulatory function of MBNL1 binding to its own transcript. Our data indicate that MBNLs induce skipping of e1 from precursor MBNL1 mRNA and that e1 exclusion may impact transcript association with polysomes and translation. Furthermore, e1-deficient protein isoform lacking the first two zinc fingers is highly unstable and its EGFP fusion protein has severely compromised splicing activity. We also show that MBNL1 can be transcribed from three different promoters and that the transcription initiation site determines the mode of e1 regulation. Taken together, we demonstrate that MBNL proteins control steady-state levels of MBNL1 through an interaction with e1 in its precursor mRNA. Insights from our study open a new avenue in therapies against DM based on manipulation of the transcription initiation site and e1 splicing of MBNL1 mRNA.
Collapse
Affiliation(s)
- Patryk Konieczny
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Ewa Stepniak-Konieczna
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Katarzyna Taylor
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Lukasz J Sznajder
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Krzysztof Sobczak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| |
Collapse
|
61
|
Thomas JD, Sznajder ŁJ, Bardhi O, Aslam FN, Anastasiadis ZP, Scotti MM, Nishino I, Nakamori M, Wang ET, Swanson MS. Disrupted prenatal RNA processing and myogenesis in congenital myotonic dystrophy. Genes Dev 2017; 31:1122-1133. [PMID: 28698297 PMCID: PMC5538435 DOI: 10.1101/gad.300590.117] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023]
Abstract
Thomas et al. demonstrate that RNA misprocessing is a major pathogenic factor in congenital myotonic dystrophy and provide novel mouse models to further examine roles for cotranscriptional/post-transcriptional gene regulation during tissue development. Myotonic dystrophy type 1 (DM1) is a CTG microsatellite expansion (CTGexp) disorder caused by expression of CUGexp RNAs. These mutant RNAs alter the activities of RNA processing factors, including MBNL proteins, leading to re-expression of fetal isoforms in adult tissues and DM1 pathology. While this pathogenesis model accounts for adult-onset disease, the molecular basis of congenital DM (CDM) is unknown. Here, we test the hypothesis that disruption of developmentally regulated RNA alternative processing pathways contributes to CDM disease. We identify prominent alternative splicing and polyadenylation abnormalities in infant CDM muscle, and, although most are also misregulated in adult-onset DM1, dysregulation is significantly more severe in CDM. Furthermore, analysis of alternative splicing during human myogenesis reveals that CDM-relevant exons undergo prenatal RNA isoform transitions and are predicted to be disrupted by CUGexp-associated mechanisms in utero. To test this possibility and the contribution of MBNLs to CDM pathogenesis, we generated mouse Mbnl double (Mbnl1; Mbnl2) and triple (Mbnl1; Mbnl2; Mbnl3) muscle-specific knockout models that recapitulate the congenital myopathy, gene expression, and spliceopathy defects characteristic of CDM. This study demonstrates that RNA misprocessing is a major pathogenic factor in CDM and provides novel mouse models to further examine roles for cotranscriptional/post-transcriptional gene regulation during development.
Collapse
Affiliation(s)
- James D Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Łukasz J Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Olgert Bardhi
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Faaiq N Aslam
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Zacharias P Anastasiadis
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Marina M Scotti
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Center of Neurology and Psychiatry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eric T Wang
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
62
|
Normal and altered pre-mRNA processing in the DMD gene. Hum Genet 2017; 136:1155-1172. [DOI: 10.1007/s00439-017-1820-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/02/2017] [Indexed: 12/11/2022]
|
63
|
Xie S, Chen L, Zhang X, Liu X, Chen Y, Mo D. An integrated analysis revealed different microRNA-mRNA profiles during skeletal muscle development between Landrace and Lantang pigs. Sci Rep 2017; 7:2516. [PMID: 28566753 PMCID: PMC5451474 DOI: 10.1038/s41598-017-02558-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/13/2017] [Indexed: 11/12/2022] Open
Abstract
Pigs supply vital dietary proteins for human consumption, and their economic value depends largely on muscle production. MicroRNAs are known to play important roles in skeletal muscle development. However, their relationship to distinct muscle production between pig breeds remains unknown. Here, we performed an integrated analysis of microRNA-mRNA expression profiles for Landrace (LR, lean) pigs and the Chinese indigenous Lantang pig (LT, lard-type) during 8 stages of skeletal muscle developmental, including at 35, 49, 63, 77 dpc (days post coitum) and 2, 28, 90, 180 dpn (days postnatal). As differentially expressed-miRNA expression profiles can be well classified into two clusters by PCA analysis, we grouped the embryonic stages as G1 and the postnatal stages as G2. A total of 203 genes were predicted miRNA targets, and a STEM analysis showed distinct expression patterns between G1 and G2 in both breeds based on their transcriptomic data. Furthermore, a STRING analysis predicted interactions between 22 genes and 35 miRNAs, including some crucial myogenic factors and myofibrillar genes. Thus, it can be reasonably speculated that myogenic miRNAs may regulate myofibrillar genes in myofiber formation during embryonic stages and muscle hypertrophy during postnatal stages, leading to distinct differences in muscle production between breeds.
Collapse
Affiliation(s)
- Shuihua Xie
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
- General Station of Animal Husbandry Technology Extension, Department of Agriculture of Guangdong Province, Guangzhou, 510500, Guangdong, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Xumeng Zhang
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
64
|
Niba ETE, Yamanaka R, Rani AQM, Awano H, Matsumoto M, Nishio H, Matsuo M. DMD transcripts in CRL-2061 rhabdomyosarcoma cells show high levels of intron retention by intron-specific PCR amplification. Cancer Cell Int 2017; 17:58. [PMID: 28546788 PMCID: PMC5442858 DOI: 10.1186/s12935-017-0428-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The DMD gene encoding dystrophin is mutated in Duchenne muscular dystrophy, a fatal progressive muscle wasting disease. DMD has also been shown to act as a tumor suppressor gene. Rhabdomyosarcoma (RMS) is a mesodermal sarcoma that shares characteristics of skeletal muscle precursors. Products of the DMD gene in RMS have not yet been fully clarified. Here, DMD products were analyzed in CRL-2061 cells established from alveolar RMS. METHODS The 14-kb long DMD cDNA was PCR amplified as 20 separated fragments, as were nine short intron regions. Dystrophin was analyzed by Western blotting using an antibody against the C-terminal region of dystrophin. RESULTS Sixteen of the 20 DMD cDNA fragments could be amplified from CRL-2061 cells as muscle cDNA. Three fragments included aberrant gene products, including one in which exon 71 was omitted and one each with retention of introns 40 and 58. In one fragment, extending from exon 70 to 79, no normally spliced product was obtained. Rather, six alternatively spliced products were identified, including a new product deleting exon 73, with the most abundant product showing deletion of exon 78. Although dystrophin expression was expected in CRL-2061 cells, western blotting of cell lysates showed no evidence of dystrophin, suggesting that translation of full-length DMD mRNA was inhibited by intron retention that generated a premature stop codon. Intron specific PCR amplification of nine short introns, showed retention of introns 40, 58, and 70, which constituted about 60, 25 and 9%, respectively, of the total PCR amplified products. The most abundant DMD transcript contained two abnormalities, intron 40 retention and exon 78 skipping. CONCLUSIONS Intron-specific PCR amplification showed that DMD transcripts contained high levels of introns 40, 58 and 70. Retention of these introns may have been responsible for the lack of dystrophin expression by CRL-2061 cells, thereby abolishing the tumor suppressor activity of dystrophin.
Collapse
Affiliation(s)
- Emma Tabe Eko Niba
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| | - Ryo Yamanaka
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| | - Abdul Qawee Mahyoob Rani
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan.,Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Masaaki Matsumoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Hisahide Nishio
- Department of Community Medicine and Social Healthcare Sciences, Kobe University Graduate School of Medicine, Chuo, Kobe, 6500017 Japan
| | - Masafumi Matsuo
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, 518 Arise, Ikawadani, Nishi, Kobe, 6512180 Japan
| |
Collapse
|
65
|
Zhang B, Zhang ZG, Yan X, Wang XX, Zhao H, Guo J, Feng JY, Long YZ. Chitosan nanostructures by in situ electrospinning for high-efficiency PM2.5 capture. NANOSCALE 2017; 9:4154-4161. [PMID: 28282101 DOI: 10.1039/c6nr09525a] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Nanofiber-based air filters and electrostatic precipitation have stimulated considerable interest because of their high-efficiency for PM2.5 capture. In this paper, we introduce a new method of in situ electrospinning (e-spinning) of nanostructures into a polluted enclosed space to efficiently clean the air. From the comparisons of different polymer precursors and different PM2.5 capture techniques, it can be seen that in situ e-spinning of chitosan aqueous solution into the air exhibits the best PM2.5 capture efficiency, which may be attributed to the stronger polarity of chitosan and the synergistic effect of the strong electrostatic adsorption and surface adhesion of the electrospun (e-spun) nanofibers. A removal rate as high as 3.7 μg m-3 s-1 was obtained using this technology with a high efficiency of more than 95% PM2.5 capture. The results obtained from a field test in a smoking room (∼5 × 6 × 3 m3) are still in great agreement with those obtained in an experimental box (∼25 × 30 × 35 cm3). More importantly, chitosan is non-toxic and biodegradable, and is harmless to human health when used as a precursor for in situ e-spinning for PM2.5 capture.
Collapse
Affiliation(s)
- Bin Zhang
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Zhi-Guang Zhang
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Xu Yan
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China. and Industrial Research Institute of Nonwovens & Technical Textiles, Qingdao University, Qingdao 266071, China
| | - Xiao-Xiong Wang
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Hui Zhao
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Jia Guo
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Ji-Yong Feng
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China.
| | - Yun-Ze Long
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China. and Industrial Research Institute of Nonwovens & Technical Textiles, Qingdao University, Qingdao 266071, China
| |
Collapse
|
66
|
Arandel L, Polay Espinoza M, Matloka M, Bazinet A, De Dea Diniz D, Naouar N, Rau F, Jollet A, Edom-Vovard F, Mamchaoui K, Tarnopolsky M, Puymirat J, Battail C, Boland A, Deleuze JF, Mouly V, Klein AF, Furling D. Immortalized human myotonic dystrophy muscle cell lines to assess therapeutic compounds. Dis Model Mech 2017; 10:487-497. [PMID: 28188264 PMCID: PMC5399563 DOI: 10.1242/dmm.027367] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/05/2017] [Indexed: 01/20/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant neuromuscular diseases caused by microsatellite expansions and belong to the family of RNA-dominant disorders. Availability of cellular models in which the DM mutation is expressed within its natural context is essential to facilitate efforts to identify new therapeutic compounds. Here, we generated immortalized DM1 and DM2 human muscle cell lines that display nuclear RNA aggregates of expanded repeats, a hallmark of myotonic dystrophy. Selected clones of DM1 and DM2 immortalized myoblasts behave as parental primary myoblasts with a reduced fusion capacity of immortalized DM1 myoblasts when compared with control and DM2 cells. Alternative splicing defects were observed in differentiated DM1 muscle cell lines, but not in DM2 lines. Splicing alterations did not result from differentiation delay because similar changes were found in immortalized DM1 transdifferentiated fibroblasts in which myogenic differentiation has been forced by overexpression of MYOD1. As a proof-of-concept, we show that antisense approaches alleviate disease-associated defects, and an RNA-seq analysis confirmed that the vast majority of mis-spliced events in immortalized DM1 muscle cells were affected by antisense treatment, with half of them significantly rescued in treated DM1 cells. Immortalized DM1 muscle cell lines displaying characteristic disease-associated molecular features such as nuclear RNA aggregates and splicing defects can be used as robust readouts for the screening of therapeutic compounds. Therefore, immortalized DM1 and DM2 muscle cell lines represent new models and tools to investigate molecular pathophysiological mechanisms and evaluate the in vitro effects of compounds on RNA toxicity associated with myotonic dystrophy mutations. Summary: Myotonic dystrophy muscle cell models displaying characteristic disease-associated molecular features can be used to investigate molecular pathophysiological mechanisms and evaluate therapeutic approaches.
Collapse
Affiliation(s)
- Ludovic Arandel
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Micaela Polay Espinoza
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Magdalena Matloka
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Audrey Bazinet
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Damily De Dea Diniz
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Naïra Naouar
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Frédérique Rau
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Arnaud Jollet
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Frédérique Edom-Vovard
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Kamel Mamchaoui
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Mark Tarnopolsky
- McMaster University Medical Center, Departments of Pediatrics and Medicine, 1200 Main St W., Hamilton, Ontario, Canada, L8N 3Z5
| | - Jack Puymirat
- CHU de Quebec, site Enfant-Jésus, Université Laval, Québec, Canada G1J 1Z4
| | - Christophe Battail
- Centre National de Génotypage, Institut de Génomique, CEA, 91000 Evry, France
| | - Anne Boland
- Centre National de Génotypage, Institut de Génomique, CEA, 91000 Evry, France
| | | | - Vincent Mouly
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Arnaud F Klein
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| | - Denis Furling
- Sorbonne Universités UPMC Univ Paris 06, INSERM, CNRS, Centre de Recherche en Myologie, Institut de Myologie, GH Pitié-Salpêtrière, Paris 75013, France
| |
Collapse
|
67
|
Dystrophin Dp71 Isoforms Are Differentially Expressed in the Mouse Brain and Retina: Report of New Alternative Splicing and a Novel Nomenclature for Dp71 Isoforms. Mol Neurobiol 2017; 55:1376-1386. [PMID: 28127699 DOI: 10.1007/s12035-017-0405-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022]
Abstract
Multiple dystrophin Dp71 isoforms have been identified in rats, mice, and humans and in several cell line models. These Dp71 isoforms are produced by the alternative splicing of exons 71 to 74 and 78 and intron 77. Three main groups of Dp71 proteins are defined based on their C-terminal specificities: Dp71d, Dp71f, and Dp71e. Dp71 is highly expressed in the brain and retina; however, the specific isoforms present in these tissues have not been determined to date. In this work, we explored the expression of Dp71 isoforms in the mouse brain and retina using RT-PCR assays followed by the cloning of PCR products into the pGEM-T Easy vector, which was used to transform DH5α cells. Dp71-positive colonies were later analyzed by PCR multiplex and DNA sequencing to determine the alternative splicing. We thus demonstrated the expression of Dp71 transcripts corresponding to Dp71, Dp71a, Dp71c, Dp71b, Dp71ab, Dp71 Δ110, and novel Dp71 isoforms spliced in exon 74; 71 and 74; 71, 73 and 74; and 74 and 78, which we named Dp71d Δ74 , Dp71d Δ71,74 , Dp71d Δ71,73-74 , and Dp71f Δ74 , respectively. Additionally, we demonstrated that the Dp71d group of isoforms is highly expressed in the brain, while the Dp71f group predominates in the retina, at both the cDNA and protein levels. These findings suggest that distinct Dp71 isoforms may play different roles in the brain and retina.
Collapse
|
68
|
Targeted RNA-Seq profiling of splicing pattern in the DMD gene: exons are mostly constitutively spliced in human skeletal muscle. Sci Rep 2017; 7:39094. [PMID: 28045018 PMCID: PMC5206723 DOI: 10.1038/srep39094] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 11/17/2016] [Indexed: 02/08/2023] Open
Abstract
We have analysed the splicing pattern of the human Duchenne Muscular Dystrophy (DMD) NB transcript in normal skeletal muscle. To achieve depth of coverage required for the analysis of this lowly expressed gene in muscle, we designed a targeted RNA-Seq procedure that combines amplification of the full-length 11.3 kb DMD cDNA sequence and 454 sequencing technology. A high and uniform coverage of the cDNA sequence was obtained that allowed to draw up a reliable inventory of the physiological alternative splicing events in the muscular DMD transcript. In contrast to previous assumptions, we evidenced that most of the 79 DMD exons are constitutively spliced in skeletal muscle. Only a limited number of 12 alternative splicing events were identified, all present at a very low level. These include previously known exon skipping events but also newly described pseudoexon inclusions and alternative 3′ splice sites, of which one is the first functional NAGNAG splice site reported in the DMD gene. This study provides the first RNA-Seq-based reference of DMD splicing pattern in skeletal muscle and reports on an experimental procedure well suited to detect condition-specific differences in this low abundance transcript that may prove useful for diagnostic, research or RNA-based therapeutic applications.
Collapse
|
69
|
Zhu C, Chen Z, Guo W. Pre-mRNA mis-splicing of sarcomeric genes in heart failure. Biochim Biophys Acta Mol Basis Dis 2016; 1863:2056-2063. [PMID: 27825848 DOI: 10.1016/j.bbadis.2016.11.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/11/2016] [Accepted: 11/01/2016] [Indexed: 12/01/2022]
Abstract
Pre-mRNA splicing is an important biological process that allows production of multiple proteins from a single gene in the genome, and mainly contributes to protein diversity in eukaryotic organisms. Alternative splicing is commonly governed by RNA binding proteins to meet the ever-changing demands of the cell. However, the mis-splicing may lead to human diseases. In the heart of human, mis-regulation of alternative splicing has been associated with heart failure. In this short review, we focus on alternative splicing of sarcomeric genes and review mis-splicing related heart failure with relatively well studied Sarcomeric genes and splicing mechanisms with identified regulatory factors. The perspective of alternative splicing based therapeutic strategies in heart failure has also been discussed.
Collapse
Affiliation(s)
- Chaoqun Zhu
- Animal Science, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, USA
| | - Zhilong Chen
- Animal Science, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, USA; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wei Guo
- Animal Science, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
70
|
Splicing misregulation of SCN5A contributes to cardiac-conduction delay and heart arrhythmia in myotonic dystrophy. Nat Commun 2016; 7:11067. [PMID: 27063795 PMCID: PMC4831019 DOI: 10.1038/ncomms11067] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 02/16/2016] [Indexed: 12/11/2022] Open
Abstract
Myotonic dystrophy (DM) is caused by the expression of mutant RNAs containing expanded CUG repeats that sequester muscleblind-like (MBNL) proteins, leading to alternative splicing changes. Cardiac alterations, characterized by conduction delays and arrhythmia, are the second most common cause of death in DM. Using RNA sequencing, here we identify novel splicing alterations in DM heart samples, including a switch from adult exon 6B towards fetal exon 6A in the cardiac sodium channel, SCN5A. We find that MBNL1 regulates alternative splicing of SCN5A mRNA and that the splicing variant of SCN5A produced in DM presents a reduced excitability compared with the control adult isoform. Importantly, reproducing splicing alteration of Scn5a in mice is sufficient to promote heart arrhythmia and cardiac-conduction delay, two predominant features of myotonic dystrophy. In conclusion, misregulation of the alternative splicing of SCN5A may contribute to a subset of the cardiac dysfunctions observed in myotonic dystrophy. Patients with myotonic dystrophy (MD) suffer from severe cardiac issues of unknown aetiology. Freyermuth et al. show that fatal changes in cardiac electrophysiological properties in humans and mice with MD may arise from misregulation of the alternative splicing of the cardiac Na+ channel SCN5A transcript, resulting in expression of its fetal form.
Collapse
|
71
|
Massouridès E, Polentes J, Mangeot PE, Mournetas V, Nectoux J, Deburgrave N, Nusbaum P, Leturcq F, Popplewell L, Dickson G, Wein N, Flanigan KM, Peschanski M, Chelly J, Pinset C. Dp412e: a novel human embryonic dystrophin isoform induced by BMP4 in early differentiated cells. Skelet Muscle 2015; 5:40. [PMID: 26568816 PMCID: PMC4644319 DOI: 10.1186/s13395-015-0062-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/21/2015] [Indexed: 01/09/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a devastating X-linked recessive genetic myopathy. DMD physiopathology is still not fully understood and a prenatal onset is suspected but difficult to address. Methods The bone morphogenetic protein 4 (BMP4) is a critical signaling molecule involved in mesoderm commitment. Human induced pluripotent stem cells (hiPSCs) from DMD and healthy individuals and human embryonic stem cells (hESCs) treated with BMP4 allowed us to model the early steps of myogenesis in normal and DMD contexts. Results Unexpectedly, 72h following BMP4 treatment, a new long DMD transcript was detected in all tested hiPSCs and hESCs, at levels similar to that found in adult skeletal muscle. This novel transcript named “Dp412e” has a specific untranslated first exon which is conserved only in a sub-group of anthropoids including human. The corresponding novel dystrophin protein of 412-kiloDalton (kDa), characterized by an N-terminal-truncated actin-binding domain, was detected in normal BMP4-treated hiPSCs/hESCs and in embryoid bodies. Finally, using a phosphorodiamidate morpholino oligomer (PMO) targeting the DMD exon 53, we demonstrated the feasibility of exon skipping validation with this BMP4-inducible hiPSCs model. Conclusions In this study, the use of hiPSCs to analyze early phases of human development in normal and DMD contexts has led to the discovery of an embryonic 412 kDa dystrophin isoform. Deciphering the regulation process(es) and the function(s) associated to this new isoform can contribute to a better understanding of the DMD physiopathology and potential developmental defects. Moreover, the simple and robust BMP4-inducible model highlighted here, providing large amount of a long DMD transcript and the corresponding protein in only 3 days, is already well-adapted to high-throughput and high-content screening approaches. Therefore, availability of this powerful cell platform can accelerate the development, validation and improvement of DMD genetic therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0062-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jérôme Polentes
- I-STEM, CECS, Génopôle-Campus 1, 5 rue Henri Desbruères, 91030 Evry, Cedex France
| | - Philippe-Emmanuel Mangeot
- CIRI, International Center for Infectiology Research, Université de Lyon, Lyon, France ; Inserm, U1111, Lyon, France ; CNRS, UMR5308, Lyon, France ; Ecole Normale Supérieure de Lyon, Lyon, France ; Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
| | | | - Juliette Nectoux
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Nathalie Deburgrave
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Patrick Nusbaum
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - France Leturcq
- Service de Biochimie et Génétique Moléculaire, HUPC Hôpital Cochin, Paris, France
| | - Linda Popplewell
- School of Biological Sciences, Royal Holloway-University of London, Surrey, TW20 0EX UK
| | - George Dickson
- School of Biological Sciences, Royal Holloway-University of London, Surrey, TW20 0EX UK
| | - Nicolas Wein
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205 USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205 USA
| | - Marc Peschanski
- UEVE U861, 91030 Evry, France ; Inserm U861, 91030 Evry, France
| | - Jamel Chelly
- IGBMC-CNRS UMR7104/Inserm U964, 67404 Illkirch, Cedex France
| | | |
Collapse
|