1
|
El Boujnouni N, van der Bent ML, Willemse M, ’t Hoen PA, Brock R, Wansink DG. Block or degrade? Balancing on- and off-target effects of antisense strategies against transcripts with expanded triplet repeats in DM1. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:622-636. [PMID: 37200862 PMCID: PMC10185704 DOI: 10.1016/j.omtn.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/13/2023] [Indexed: 05/20/2023]
Abstract
Antisense oligonucleotide (ASO) therapies for myotonic dystrophy type 1 (DM1) are based on elimination of transcripts containing an expanded repeat or inhibition of sequestration of RNA-binding proteins. This activity is achievable by both degradation of expanded transcripts and steric hindrance, although it is unknown which approach is superior. We compared blocking ASOs with RNase H-recruiting gapmers of equivalent chemistries. Two DMPK target sequences were selected: the triplet repeat and a unique sequence upstream thereof. We assessed ASO effects on transcript levels, ribonucleoprotein foci and disease-associated missplicing, and performed RNA sequencing to investigate on- and off-target effects. Both gapmers and the repeat blocker led to significant DMPK knockdown and a reduction in (CUG)exp foci. However, the repeat blocker was more effective in MBNL1 protein displacement and had superior efficiency in splicing correction at the tested dose of 100 nM. By comparison, on a transcriptome level, the blocking ASO had the fewest off-target effects. In particular, the off-target profile of the repeat gapmer asks for cautious consideration in further therapeutic development. Altogether, our study demonstrates the importance of evaluating both on-target and downstream effects of ASOs in a DM1 context, and provides guiding principles for safe and effective targeting of toxic transcripts.
Collapse
Affiliation(s)
- Najoua El Boujnouni
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - M. Leontien van der Bent
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Marieke Willemse
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Peter A.C. ’t Hoen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Roland Brock
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 293, Bahrain
- Corresponding author Roland Brock, Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| | - Derick G. Wansink
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Corresponding author Derick G. Wansink, Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
2
|
Liu J, Guo ZN, Yan XL, Yang Y, Huang S. Brain Pathogenesis and Potential Therapeutic Strategies in Myotonic Dystrophy Type 1. Front Aging Neurosci 2021; 13:755392. [PMID: 34867280 PMCID: PMC8634727 DOI: 10.3389/fnagi.2021.755392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy that affects multiple systems including the muscle and heart. The mutant CTG expansion at the 3'-UTR of the DMPK gene causes the expression of toxic RNA that aggregate as nuclear foci. The foci then interfere with RNA-binding proteins, affecting hundreds of mis-spliced effector genes, leading to aberrant alternative splicing and loss of effector gene product functions, ultimately resulting in systemic disorders. In recent years, increasing clinical, imaging, and pathological evidence have indicated that DM1, though to a lesser extent, could also be recognized as true brain diseases, with more and more researchers dedicating to develop novel therapeutic tools dealing with it. In this review, we summarize the current advances in the pathogenesis and pathology of central nervous system (CNS) deficits in DM1, intervention measures currently being investigated are also highlighted, aiming to promote novel and cutting-edge therapeutic investigations.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Shuo Huang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
3
|
Ausems CRM, Raaijmakers RHL, van den Broek WJAA, Willemse M, van Engelen BGM, Wansink DG, van Bokhoven H. Intrinsic Myogenic Potential of Skeletal Muscle-Derived Pericytes from Patients with Myotonic Dystrophy Type 1. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:120-132. [PMID: 31649961 PMCID: PMC6804802 DOI: 10.1016/j.omtm.2019.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022]
Abstract
Pericytes are multipotent, vessel-associated progenitors that exhibit high proliferative capacity, can cross the blood-muscle barrier, and have the ability to home to muscle tissue and contribute to myogenesis. Consequently, pericyte-based therapies hold great promise for muscular dystrophies. A complex multi-system disorder exhibiting muscular dystrophy for which pericytes might be a valuable cell source is myotonic dystrophy type 1 (DM1). DM1 is caused by an unstable (CTG)n repeat in the DMPK gene and characterized by skeletal muscle weakness, muscle wasting, and myotonia. We have successfully isolated alkaline phosphatase-positive pericytes from skeletal muscle of DM1 patients and a transgenic mouse model. Intranuclear (CUG)n RNA foci, a pathogenic DM1 hallmark, were identified in human and mouse pericytes. Notably, pericytes from DM1 patients maintained similar growth parameters and innate myogenic characteristics in vitro compared to cells from unaffected controls. Our in vitro results thus demonstrate the potential of pericytes to ameliorate muscle features in DM1 in a therapeutic setting.
Collapse
Affiliation(s)
- Cornelia Rosanne Maria Ausems
- Department of Human Genetics, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6525 GA Nijmegen, the Netherlands.,Department of Neurology, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6500 HB Nijmegen, the Netherlands.,Department of Cell Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, the Netherlands
| | - Renée Henrica Lamberta Raaijmakers
- Department of Human Genetics, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6525 GA Nijmegen, the Netherlands.,Department of Neurology, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6500 HB Nijmegen, the Netherlands.,Department of Cell Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, the Netherlands
| | | | - Marieke Willemse
- Department of Cell Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, the Netherlands
| | - Baziel Gerardus Maria van Engelen
- Department of Neurology, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6500 HB Nijmegen, the Netherlands
| | - Derick Gert Wansink
- Department of Cell Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, the Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud University Medical Center, Donders lnstitute for Brain Cognition and Behavior, 6525 GA Nijmegen, the Netherlands
| |
Collapse
|
4
|
van der Bent ML, Paulino da Silva Filho O, Willemse M, Hällbrink M, Wansink DG, Brock R. The nuclear concentration required for antisense oligonucleotide activity in myotonic dystrophy cells. FASEB J 2019; 33:11314-11325. [PMID: 31311315 DOI: 10.1096/fj.201900263r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antisense oligonucleotides (ASOs) are a promising class of therapeutics that are starting to emerge in the clinic. Determination of intracellular concentrations required for biologic effects and identification of effective delivery vehicles are crucial for understanding the mode of action and required dosing. Here, we investigated which nuclear oligonucleotide concentration is needed for a therapeutic effect for a triplet repeat-targeting ASO in a muscle cell model of myotonic dystrophy type 1 (DM1). For cellular delivery, ASOs were complexed into nanoparticles using the cationic cell-penetrating peptides nona-arginine and PepFect14 (PF14). Although both peptides facilitated uptake, only PF14 led to a dose-dependent correction of disease-typical abnormal splicing. In line with this observation, time-lapse confocal microscopy demonstrated that only PF14 mediated translocation of the ASOs to the nucleus, which is the main site of action. Through fluorescence lifetime imaging, we could distinguish intact oligonucleotide from free fluorophore, showing that PF14 also shielded the ASOs from degradation. Finally, we employed a combination of live-cell fluorescence correlation spectroscopy and immunofluorescence microscopy and demonstrated that intranuclear blocking-type oligonucleotide concentrations in the upper nanomolar range were required to dissolve nuclear muscleblind-like protein 1 foci, a hallmark of DM1. Our findings have important implications for the clinical use of ASOs in DM1 and provide a basis for further research on other types of ASOs.-Van der Bent, M. L., Paulino da Silva Filho, O., Willemse, M., Hällbrink, M., Wansink, D. G., Brock, R. The nuclear concentration required for antisense oligonucleotide activity in myotonic dystrophy cells.
Collapse
Affiliation(s)
- M Leontien van der Bent
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Omar Paulino da Silva Filho
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) Foundation, Ministry of Education of Brazil, Brasilia, Brazil
| | - Marieke Willemse
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mattias Hällbrink
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Heskamp L, van Nimwegen M, Ploegmakers MJ, Bassez G, Deux JF, Cumming SA, Monckton DG, van Engelen BGM, Heerschap A. Lower extremity muscle pathology in myotonic dystrophy type 1 assessed by quantitative MRI. Neurology 2019; 92:e2803-e2814. [PMID: 31118244 PMCID: PMC6598795 DOI: 10.1212/wnl.0000000000007648] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/07/2019] [Indexed: 01/08/2023] Open
Abstract
Objective To determine the value of quantitative MRI in providing imaging biomarkers for disease in 20 different upper and lower leg muscles of patients with myotonic dystrophy type 1 (DM1). Methods We acquired images covering these muscles in 33 genetically and clinically well-characterized patients with DM1 and 10 unaffected controls. MRIs were recorded with a Dixon method to determine muscle fat fraction, muscle volume, and contractile muscle volume, and a multi-echo spin-echo sequence was used to determine T2 water relaxation time (T2water), reflecting putative edema. Results Muscles in patients with DM1 had higher fat fractions than muscles of controls (15.6 ± 11.1% vs 3.7 ± 1.5%). In addition, patients had smaller muscle volumes (902 ± 232 vs 1,097 ± 251 cm3), smaller contractile muscle volumes (779 ± 247 vs 1,054 ± 246 cm3), and increased T2water (33.4 ± 1.0 vs 31.9 ± 0.6 milliseconds), indicating atrophy and edema, respectively. Lower leg muscles were affected most frequently, especially the gastrocnemius medialis and soleus. Distribution of fat content per muscle indicated gradual fat infiltration in DM1. Between-patient variation in fat fraction was explained by age (≈45%), and another ≈14% was explained by estimated progenitor CTG repeat length (r2 = 0.485) and somatic instability (r2 = 0.590). Fat fraction correlated with the 6-minute walk test (r = −0.553) and muscular impairment rating scale (r = 0.537) and revealed subclinical muscle involvement. Conclusion This cross-sectional quantitative MRI study of 20 different lower extremity muscles in patients with DM1 revealed abnormal values for muscle fat fraction, volume, and T2water, which therefore may serve as objective biomarkers to assess disease state of skeletal muscles in these patients. ClinicalTrials.gov identifier NCT02118779.
Collapse
Affiliation(s)
- Linda Heskamp
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| | - Marlies van Nimwegen
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Marieke J Ploegmakers
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Guillaume Bassez
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Jean-Francois Deux
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Sarah A Cumming
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Darren G Monckton
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Baziel G M van Engelen
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Arend Heerschap
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
6
|
Furuta N, Tsukagoshi S, Hirayanagi K, Ikeda Y. Suppression of the yeast elongation factor Spt4 ortholog reduces expanded SCA36 GGCCUG repeat aggregation and cytotoxicity. Brain Res 2019; 1711:29-40. [DOI: 10.1016/j.brainres.2018.12.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
|
7
|
Dastidar S, Ardui S, Singh K, Majumdar D, Nair N, Fu Y, Reyon D, Samara E, Gerli MF, Klein AF, De Schrijver W, Tipanee J, Seneca S, Tulalamba W, Wang H, Chai Y, In’t Veld P, Furling D, Tedesco F, Vermeesch JR, Joung JK, Chuah MK, VandenDriessche T. Efficient CRISPR/Cas9-mediated editing of trinucleotide repeat expansion in myotonic dystrophy patient-derived iPS and myogenic cells. Nucleic Acids Res 2018; 46:8275-8298. [PMID: 29947794 PMCID: PMC6144820 DOI: 10.1093/nar/gky548] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 12/17/2022] Open
Abstract
CRISPR/Cas9 is an attractive platform to potentially correct dominant genetic diseases by gene editing with unprecedented precision. In the current proof-of-principle study, we explored the use of CRISPR/Cas9 for gene-editing in myotonic dystrophy type-1 (DM1), an autosomal-dominant muscle disorder, by excising the CTG-repeat expansion in the 3'-untranslated-region (UTR) of the human myotonic dystrophy protein kinase (DMPK) gene in DM1 patient-specific induced pluripotent stem cells (DM1-iPSC), DM1-iPSC-derived myogenic cells and DM1 patient-specific myoblasts. To eliminate the pathogenic gain-of-function mutant DMPK transcript, we designed a dual guide RNA based strategy that excises the CTG-repeat expansion with high efficiency, as confirmed by Southern blot and single molecule real-time (SMRT) sequencing. Correction efficiencies up to 90% could be attained in DM1-iPSC as confirmed at the clonal level, following ribonucleoprotein (RNP) transfection of CRISPR/Cas9 components without the need for selective enrichment. Expanded CTG repeat excision resulted in the disappearance of ribonuclear foci, a quintessential cellular phenotype of DM1, in the corrected DM1-iPSC, DM1-iPSC-derived myogenic cells and DM1 myoblasts. Consequently, the normal intracellular localization of the muscleblind-like splicing regulator 1 (MBNL1) was restored, resulting in the normalization of splicing pattern of SERCA1. This study validates the use of CRISPR/Cas9 for gene editing of repeat expansions.
Collapse
Affiliation(s)
- Sumitava Dastidar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Simon Ardui
- Department of Human Genetics, University of Leuven, Leuven 3000, Belgium
| | - Kshitiz Singh
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Debanjana Majumdar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Nisha Nair
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Yanfang Fu
- Molecular Pathology Unit, Center for Cancer Research and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Deepak Reyon
- Molecular Pathology Unit, Center for Cancer Research and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Ermira Samara
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Mattia F M Gerli
- Department of Cell and Developmental Biology, University College London, London WC1E6DE, UK
| | - Arnaud F Klein
- Sorbonne Universités, INSERM, Association Institute de Myologie, Center de Recherche en Myologie, F-75013 , France
| | - Wito De Schrijver
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Jaitip Tipanee
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Sara Seneca
- Research Group Reproduction and Genetics (REGE), Center for Medical Genetics, UZ Brussels, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Warut Tulalamba
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Hui Wang
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Yoke Chin Chai
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Peter In’t Veld
- Department of Pathology, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Denis Furling
- Sorbonne Universités, INSERM, Association Institute de Myologie, Center de Recherche en Myologie, F-75013 , France
| | | | - Joris R Vermeesch
- Department of Human Genetics, University of Leuven, Leuven 3000, Belgium
| | - J Keith Joung
- Molecular Pathology Unit, Center for Cancer Research and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA02129, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Marinee K Chuah
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
- Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven 3000, Belgium
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, Brussels 1090, Belgium
- Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven 3000, Belgium
| |
Collapse
|
8
|
Coote DJ, Davis MR, Cabrera M, Needham M, Laing NG, Nowak KJ. Clinical Utility Gene Card for: autosomal dominant myotonia congenita (Thomsen Disease). Eur J Hum Genet 2018; 26:1072-1077. [PMID: 29695755 DOI: 10.1038/s41431-017-0065-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 09/28/2017] [Accepted: 11/23/2017] [Indexed: 11/09/2022] Open
Affiliation(s)
- David J Coote
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia; and Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, WA, 6009, Australia
| | - Mark R Davis
- Neurogenetics Laboratory, Department of Diagnostic Genomics, PP Block, QEII Medical Centre, Nedlands, WA, 6009, Australia
| | - Macarena Cabrera
- Department of Neurology and Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, Seville, 41013, Spain
| | - Merrilee Needham
- Western Australian Neuroscience Institute, QEII Medical Centre, Nedlands, Western Australia 6009; Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch, WA, 6150, Australia
| | - Nigel G Laing
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia; and Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, WA, 6009, Australia.,Neurogenetics Laboratory, Department of Diagnostic Genomics, PP Block, QEII Medical Centre, Nedlands, WA, 6009, Australia
| | - Kristen J Nowak
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia; and Harry Perkins Institute of Medical Research, QQ Block, QEII Medical Centre, Nedlands, WA, 6009, Australia. .,School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia. .,Office of Population Health Genomics, Public and Aboriginal Health Division, Department of Health, East Perth, WA, 6004, Australia.
| |
Collapse
|
9
|
Boussaïd G, Wahbi K, Laforet P, Eymard B, Stojkovic T, Behin A, Djillali A, Orlikowski D, Prigent H, Lofaso F. Genotype and other determinants of respiratory function in myotonic dystrophy type 1. Neuromuscul Disord 2017; 28:222-228. [PMID: 29398295 DOI: 10.1016/j.nmd.2017.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/30/2017] [Accepted: 12/20/2017] [Indexed: 11/18/2022]
Abstract
New treatments are being developed for myotonic dystrophy type 1 (DM1). To evaluate their efficacy, knowledge about the natural history of respiratory dysfunction and its relationship with the genotype will be crucial. Also needed is information on factors predicting the time-course of respiratory function in DM1. Using data from 283 patients, we built a segmented linear mixed-effects regression model to assess respiratory function changes over time. Respiratory variables associated with the CTG repeat number were identified by multivariate linear regression analysis. Cox proportional-hazards regression was used to estimate hazard ratios (HRs) for starting non-invasive ventilation (NIV). Higher CTG repeat number was associated with peak cough flow impairment (p = 0.007) and with lower values for maximal inspiratory pressure (p <0.0001) and upright vital capacity. A vital capacity decline over time was associated with older age at first evaluation (p <0.0001), higher CTG repeat number (p <0.0001), and higher baseline body mass index (p = 0.0004). NIV initiation was associated with lower peak cough flow (p <0.001) after age and PaCO2 adjustment. Earlier and closer monitoring with routine peak cough flow determination in adults with congenital DM1, combined with weight control, may diminish the risk of respiratory complications and optimise other aspects of management.
Collapse
Affiliation(s)
- Ghilas Boussaïd
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, Garches, France; Association Française contre les Myopathies - Téléthon, Evry, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France.
| | - Karim Wahbi
- Service de cardiologie, Hôpital Cochin, APHP, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Pascal Laforet
- Institut de Myologie, Centre de Référence de Pathologie Neuromusculaire Paris-Est, GH Pitié-Salpêtrière, Paris, France
| | - Bruno Eymard
- Institut de Myologie, Centre de Référence de Pathologie Neuromusculaire Paris-Est, GH Pitié-Salpêtrière, Paris, France
| | - Tanya Stojkovic
- Institut de Myologie, Centre de Référence de Pathologie Neuromusculaire Paris-Est, GH Pitié-Salpêtrière, Paris, France
| | - Anthony Behin
- Institut de Myologie, Centre de Référence de Pathologie Neuromusculaire Paris-Est, GH Pitié-Salpêtrière, Paris, France
| | - Annane Djillali
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, Garches, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, Garches, France
| | - David Orlikowski
- CIC 1429, INSERM, AP-HP, Hôpital Raymond Poincaré, Garches, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Pôle de ventilation à domicile, AP-HP, Hôpital Raymond Poincaré, Garches, France
| | - Hélène Prigent
- Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Service d'Explorations Fonctionnelles Respiratoires, AP-HP, Hôpital Raymond Poincaré, Garches, France
| | - Frédéric Lofaso
- Association Française contre les Myopathies - Téléthon, Evry, France; Université de Versailles Saint Quentin en Yvelines, INSERM U1179, France; Service d'Explorations Fonctionnelles Respiratoires, AP-HP, Hôpital Raymond Poincaré, Garches, France
| |
Collapse
|
10
|
Chen JL, VanEtten DM, Fountain MA, Yildirim I, Disney MD. Structure and Dynamics of RNA Repeat Expansions That Cause Huntington's Disease and Myotonic Dystrophy Type 1. Biochemistry 2017; 56:3463-3474. [PMID: 28617590 PMCID: PMC5810133 DOI: 10.1021/acs.biochem.7b00252] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RNA repeat expansions cause a host of incurable, genetically defined diseases. The most common class of RNA repeats consists of trinucleotide repeats. These long, repeating transcripts fold into hairpins containing 1 × 1 internal loops that can mediate disease via a variety of mechanism(s) in which RNA is the central player. Two of these disorders are Huntington's disease and myotonic dystrophy type 1, which are caused by r(CAG) and r(CUG) repeats, respectively. We report the structures of two RNA constructs containing three copies of a r(CAG) [r(3×CAG)] or r(CUG) [r(3×CUG)] motif that were modeled with nuclear magnetic resonance spectroscopy and simulated annealing with restrained molecular dynamics. The 1 × 1 internal loops of r(3×CAG) are stabilized by one-hydrogen bond (cis Watson-Crick/Watson-Crick) AA pairs, while those of r(3×CUG) prefer one- or two-hydrogen bond (cis Watson-Crick/Watson-Crick) UU pairs. Assigned chemical shifts for the residues depended on the identity of neighbors or next nearest neighbors. Additional insights into the dynamics of these RNA constructs were gained by molecular dynamics simulations and a discrete path sampling method. Results indicate that the global structures of the RNA are A-form and that the loop regions are dynamic. The results will be useful for understanding the dynamic trajectory of these RNA repeats but also may aid in the development of therapeutics.
Collapse
Affiliation(s)
- Jonathan L. Chen
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Damian M. VanEtten
- Department of Chemistry and Biochemistry, State University of New York at Fredonia, Fredonia, New York 14063, United States
| | - Matthew A. Fountain
- Department of Chemistry and Biochemistry, State University of New York at Fredonia, Fredonia, New York 14063, United States
| | - Ilyas Yildirim
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| | - Matthew D. Disney
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Chemistry and Biochemistry, Florida Atlantic University, Jupiter, Florida 33458, United States
| |
Collapse
|
11
|
Li J, Matsumoto J, Bai LP, Murata A, Dohno C, Nakatani K. A Ligand That Targets CUG Trinucleotide Repeats. Chemistry 2016; 22:14881-14889. [PMID: 27573860 DOI: 10.1002/chem.201602741] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Indexed: 11/06/2022]
Abstract
The development of small molecules that can recognize specific RNA secondary and tertiary structures is currently an important research topic for developing tools to modulate gene expression and therapeutic drugs. Expanded CUG trinucleotide repeats, known as toxic RNA, capture the splicing factor MBNL1 and are causative of neurological disorder myotonic dystrophy type 1 (DM1). Herein, the rational molecular design, synthesis, and binding analysis of 2,9-diaminoalkyl-substituted 1,10-phenanthroline (DAP), which bound to CUG trinucleotide repeats, is described. The results of melting temperature (Tm ) analyses, surface plasmon resonance (SPR) assay, and electrospray spray ionization time-of-flight (ESI-TOF) mass spectrometry showed that DAP bound to r(CUG)9 but not to r(CAG)9 and r(CGG)9 . The dual luciferase assay clearly indicated DAP bound to the r(CUG)n repeat by affecting the translation in vitro.
Collapse
Affiliation(s)
- Jinxing Li
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Jun Matsumoto
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Li-Ping Bai
- State Key Laboratory of Quality Research in Chinese Medicine and, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, China
| | - Asako Murata
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Chikara Dohno
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Kazuhiko Nakatani
- Department of Regulatory Bioorganic Chemistry, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan.
| |
Collapse
|
12
|
Modified Antisense Oligonucleotides and Their Analogs in Therapy of Neuromuscular Diseases. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-34175-0_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
13
|
González ÀL, Teixidó J, Borrell JI, Estrada-Tejedor R. On the Applicability of Elastic Network Models for the Study of RNA CUG Trinucleotide Repeat Overexpansion. PLoS One 2016; 11:e0152049. [PMID: 27010216 PMCID: PMC4806922 DOI: 10.1371/journal.pone.0152049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 03/08/2016] [Indexed: 11/18/2022] Open
Abstract
Non-coding RNAs play a pivotal role in a number of diseases promoting an aberrant sequestration of nuclear RNA-binding proteins. In the particular case of myotonic dystrophy type 1 (DM1), a multisystemic autosomal dominant disease, the formation of large non-coding CUG repeats set up long-tract hairpins able to bind muscleblind-like proteins (MBNL), which trigger the deregulation of several splicing events such as cardiac troponin T (cTNT) and insulin receptor’s, among others. Evidence suggests that conformational changes in RNA are determinant for the recognition and binding of splicing proteins, molecular modeling simulations can attempt to shed light on the structural diversity of CUG repeats and to understand their pathogenic mechanisms. Molecular dynamics (MD) are widely used to obtain accurate results at atomistic level, despite being very time consuming, and they contrast with fast but simplified coarse-grained methods such as Elastic Network Model (ENM). In this paper, we assess the application of ENM (traditionally applied on proteins) for studying the conformational space of CUG repeats and compare it to conventional and accelerated MD conformational sampling. Overall, the results provided here reveal that ANM can provide useful insights into dynamic rCUG structures at a global level, and that their dynamics depend on both backbone and nucleobase fluctuations. On the other hand, ANM fail to describe local U-U dynamics of the rCUG system, which require more computationally expensive methods such as MD. Given that several limitations are inherent to both methods, we discuss here the usefulness of the current theoretical approaches for studying highly dynamic RNA systems such as CUG trinucleotide repeat overexpansions.
Collapse
Affiliation(s)
- Àlex L. González
- Grup d’Enginyeria Molecular (GEM), Institut Químic de Sarrià (IQS) – Universitat Ramon Llull (URL), Barcelona, Catalonia, 08017, Spain
| | - Jordi Teixidó
- Grup d’Enginyeria Molecular (GEM), Institut Químic de Sarrià (IQS) – Universitat Ramon Llull (URL), Barcelona, Catalonia, 08017, Spain
| | - José I. Borrell
- Grup d’Enginyeria Molecular (GEM), Institut Químic de Sarrià (IQS) – Universitat Ramon Llull (URL), Barcelona, Catalonia, 08017, Spain
| | - Roger Estrada-Tejedor
- Grup d’Enginyeria Molecular (GEM), Institut Químic de Sarrià (IQS) – Universitat Ramon Llull (URL), Barcelona, Catalonia, 08017, Spain
- * E-mail:
| |
Collapse
|
14
|
de Silva C, Subramaniam V, Price MR, Sidlow R. Transverse colonic volvulus in a 16-year-old female with congenital myotonic dystrophy: A case report. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2015. [DOI: 10.1016/j.epsc.2015.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
15
|
Khan N, Kolimi N, Rathinavelan T. Twisting right to left: A…A mismatch in a CAG trinucleotide repeat overexpansion provokes left-handed Z-DNA conformation. PLoS Comput Biol 2015; 11:e1004162. [PMID: 25876062 PMCID: PMC4395422 DOI: 10.1371/journal.pcbi.1004162] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/28/2015] [Indexed: 12/02/2022] Open
Abstract
Conformational polymorphism of DNA is a major causative factor behind several incurable trinucleotide repeat expansion disorders that arise from overexpansion of trinucleotide repeats located in coding/non-coding regions of specific genes. Hairpin DNA structures that are formed due to overexpansion of CAG repeat lead to Huntington’s disorder and spinocerebellar ataxias. Nonetheless, DNA hairpin stem structure that generally embraces B-form with canonical base pairs is poorly understood in the context of periodic noncanonical A…A mismatch as found in CAG repeat overexpansion. Molecular dynamics simulations on DNA hairpin stems containing A…A mismatches in a CAG repeat overexpansion show that A…A dictates local Z-form irrespective of starting glycosyl conformation, in sharp contrast to canonical DNA duplex. Transition from B-to-Z is due to the mechanistic effect that originates from its pronounced nonisostericity with flanking canonical base pairs facilitated by base extrusion, backbone and/or base flipping. Based on these structural insights we envisage that such an unusual DNA structure of the CAG hairpin stem may have a role in disease pathogenesis. As this is the first study that delineates the influence of a single A…A mismatch in reversing DNA helicity, it would further have an impact on understanding DNA mismatch repair. When a set of 3 nucleotides in a DNA sequence repeats beyond a certain number, it leads to incurable neurological or neuromuscular disorders. Such DNA sequences tend to form unusual DNA structures comprising of base pairing schemes different from the canonical A…T/G…C base pairs. Influence of such unusual base pairing on the overall 3-dimensional structure of DNA and its impact on the pathogenesis of disorder is not well understood. CAG repeat overexpansion that leads to Huntington’s disorder and several spinocerebellar ataxias forms noncanonical A…A base pair in between canonical C…G and G…C base pairs. However, no detailed structural information is available on the influence of an A…A mismatch on a DNA structure under any sequence context. Here, we have shown for the first time that A…A base pairing in a CAG repeat provokes the formation of left-handed Z-DNA due to the pronounced structural dissimilarity of A…A base pair with G…C base pair, leading to periodic B-Z junction. Thus, these results suggest that formation of periodic B-Z junction may be one of the molecular bases for CAG repeat instability.
Collapse
Affiliation(s)
- Noorain Khan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana State, India
| | - Narendar Kolimi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana State, India
| | | |
Collapse
|
16
|
González-Barriga A, Kranzen J, Croes HJE, Bijl S, van den Broek WJAA, van Kessel IDG, van Engelen BGM, van Deutekom JCT, Wieringa B, Mulders SAM, Wansink DG. Cell membrane integrity in myotonic dystrophy type 1: implications for therapy. PLoS One 2015; 10:e0121556. [PMID: 25799359 PMCID: PMC4370802 DOI: 10.1371/journal.pone.0121556] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 02/15/2015] [Indexed: 01/04/2023] Open
Abstract
Myotonic Dystrophy type 1 (DM1) is a multisystemic disease caused by toxic RNA from a DMPK gene carrying an expanded (CTG•CAG)n repeat. Promising strategies for treatment of DM1 patients are currently being tested. These include antisense oligonucleotides and drugs for elimination of expanded RNA or prevention of aberrant binding to RNP proteins. A significant hurdle for preclinical development along these lines is efficient systemic delivery of compounds across endothelial and target cell membranes. It has been reported that DM1 patients show elevated levels of markers of muscle damage or loss of sarcolemmal integrity in their serum and that splicing of dystrophin, an essential protein for muscle membrane structure, is abnormal. Therefore, we studied cell membrane integrity in DM1 mouse models commonly used for preclinical testing. We found that membranes in skeletal muscle, heart and brain were impermeable to Evans Blue Dye. Creatine kinase levels in serum were similar to those in wild type mice and expression of dystrophin protein was unaffected. Also in patient muscle biopsies cell surface expression of dystrophin was normal and calcium-positive fibers, indicating elevated intracellular calcium levels, were only rarely seen. Combined, our findings indicate that cells in DM1 tissues do not display compromised membrane integrity. Hence, the cell membrane is a barrier that must be overcome in future work towards effective drug delivery in DM1 therapy.
Collapse
Affiliation(s)
- Anchel González-Barriga
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Prosensa Therapeutics B.V., Leiden, The Netherlands
| | - Julia Kranzen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Huib J. E. Croes
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Suzanne Bijl
- Prosensa Therapeutics B.V., Leiden, The Netherlands
| | - Walther J. A. A. van den Broek
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Ingeborg D. G. van Kessel
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Baziel G. M. van Engelen
- Department of Neurology, Donders Centre for Neuroscience, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Susan A. M. Mulders
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Prosensa Therapeutics B.V., Leiden, The Netherlands
| | - Derick G. Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
17
|
Huang TL, Mayence A, Vanden Eynde JJ. Some non-conventional biomolecular targets for diamidines. A short survey. Bioorg Med Chem 2014; 22:1983-92. [DOI: 10.1016/j.bmc.2014.02.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/24/2022]
|
18
|
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: 4.1] [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
|
19
|
Design and analysis of effects of triplet repeat oligonucleotides in cell models for myotonic dystrophy. MOLECULAR THERAPY-NUCLEIC ACIDS 2013; 2:e81. [PMID: 23511335 PMCID: PMC3615819 DOI: 10.1038/mtna.2013.9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is caused by DM protein kinase (DMPK) transcripts containing an expanded (CUG)n repeat. Antisense oligonucleotide (AON)-mediated suppression of these mutant RNAs is considered a promising therapeutic strategy for this severe disorder. Earlier, we identified a 2'-O-methyl (2'-OMe) phosphorothioate (PT)-modified (CAG)7 oligo (PS58), which selectively silences mutant DMPK transcripts through recognition of the abnormally long (CUG)n tract. We present here a comprehensive collection of triplet repeat AONs and found that oligo length and nucleotide chemistry are important determinants for activity. For significant reduction of expanded DMPK mRNAs, a minimal length of five triplets was required. 2'-O,4'-C-ethylene-bridged nucleic acid (ENA)-modified AONs appeared not effective, probably due to lack of nuclear internalization. Selectivity for products from the expanded DMPK allele in patient myoblasts, an important requirement to minimize unwanted side effects, appeared also dependent on AON chemistry. In particular, RNase-H-dependent (CAG)n AONs did not show (CUG)n length specificity. We provide evidence that degradation of long DMPK transcripts induced by PS58-type AONs is an RNase-H independent process, does not involve oligo-intrinsic RNase activity nor does it interfere with splicing of DMPK transcripts. Our collection of triplet repeat AONs forms an important resource for further development of a safe therapy for DM1 and other unstable microsatellite diseases.Molecular Therapy-Nucleic Acids (2013) 2, e81; doi:10.1038/mtna.2013.9; published online 19 March 2013.
Collapse
|
20
|
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: 4.9] [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
|
21
|
Martínez T, Wright N, López-Fraga M, Jiménez AI, Pañeda C. Silencing human genetic diseases with oligonucleotide-based therapies. Hum Genet 2013; 132:481-93. [PMID: 23494242 DOI: 10.1007/s00439-013-1288-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 03/05/2013] [Indexed: 12/11/2022]
Abstract
RNA interference is an endogenous mechanism present in most eukaryotic cells that enables degradation of specific mRNAs. Pharmacological exploitation of this mechanism for therapeutic purposes attracted a whole amount of attention in its initial years, but was later hampered due to difficulties in delivery of the pharmacological agents to the appropriate organ or tissue. Advances in recent years have to a certain level started to address this specific issue. Genetic diseases are caused by aberrations in gene sequences or structure; these particular abnormalities are in theory easily addressable by RNAi therapeutics. Sequencing of the human genome has largely contributed to the identification of alterations responsible for genetic conditions, thus facilitating the design of compounds that can address these diseases. This review addresses the currently on-going programs with the aim of developing RNAi and other antisense compounds for the treatment of genetic conditions and the pros and cons that these products may encounter along the way. The authors have focused on those programs that have reached clinical trials or are very close to do so.
Collapse
Affiliation(s)
- Tamara Martínez
- Sylentis, PCM C/Santiago Grisolía no 2, Tres Cantos, 28760, Madrid, Spain
| | | | | | | | | |
Collapse
|
22
|
Huguet A, Medja F, Nicole A, Vignaud A, Guiraud-Dogan C, Ferry A, Decostre V, Hogrel JY, Metzger F, Hoeflich A, Baraibar M, Gomes-Pereira M, Puymirat J, Bassez G, Furling D, Munnich A, Gourdon G. Molecular, physiological, and motor performance defects in DMSXL mice carrying >1,000 CTG repeats from the human DM1 locus. PLoS Genet 2012; 8:e1003043. [PMID: 23209425 PMCID: PMC3510028 DOI: 10.1371/journal.pgen.1003043] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 09/05/2012] [Indexed: 11/22/2022] Open
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. 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.
Collapse
Affiliation(s)
- Aline Huguet
- Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Fadia Medja
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
| | - Annie Nicole
- Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Alban Vignaud
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
- Généthon, Evry, France
| | - Céline Guiraud-Dogan
- Inserm U955, Département de Neurosciences, Faculté de Médecine, Université Paris XII, Créteil, France
| | - Arnaud Ferry
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
- Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Valérie Decostre
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
| | - Jean-Yves Hogrel
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
| | - Friedrich Metzger
- F. Hoffmann-La Roche, CNS Pharma Research and Development, Basel, Switzerland
| | - Andreas Hoeflich
- Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Martin Baraibar
- UPMC Univ Paris 06, UM 76, Institut de Myologie and Inserm, U974 and CNRS, UMR7215, Paris, France
| | - Mário Gomes-Pereira
- Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Jack Puymirat
- Human Genetics Research Unit, Laval University, Québec City, Québec, Canada
| | - Guillaume Bassez
- Inserm U955, Département de Neurosciences, Faculté de Médecine, Université Paris XII, Créteil, France
| | - Denis Furling
- Institut de Myologie, Université Paris 6 UMR S974, Inserm U974, CNRS UMR 7215, GH Pitié-Salpêtrière, Paris, France
| | - Arnold Munnich
- Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Geneviève Gourdon
- Inserm U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
- * E-mail:
| |
Collapse
|
23
|
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: 335] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Cookson MR. Aging--RNA in development and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2011; 3:133-43. [PMID: 21898829 DOI: 10.1002/wrna.109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Given that RNA is involved in virtually all biological processes, it is perhaps not surprising that several RNA-binding proteins are associated with aging and with different age-related disorders. Other articles in this volume will discuss some specific examples of diseases where RNA plays a role that are also associated with aging, such as cancer and inflammation, so here I will discuss some general aspects of how RNA changes with the aging process. I will also discuss some specific examples of RNA-binding proteins that are associated with age-dependent neurological diseases as these provide an interesting framework to examine how lifetime mutations might lead to a late onset disease, although the answers to these questions are still not well understood.
Collapse
Affiliation(s)
- Mark R Cookson
- Cell Biology and Gene Expression Unit, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA.
| |
Collapse
|
25
|
Sicot G, Gourdon G, Gomes-Pereira M. Myotonic dystrophy, when simple repeats reveal complex pathogenic entities: new findings and future challenges. Hum Mol Genet 2011; 20:R116-23. [PMID: 21821673 DOI: 10.1093/hmg/ddr343] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Expanded, non-coding RNAs can exhibit a deleterious gain-of-function causing human disease through abnormal interactions with RNA-binding proteins. Myotonic dystrophy (DM), the prototypical example of an RNA-dominant disorder, is mediated by trinucleotide repeat-containing transcripts that deregulate alternative splicing. Spliceopathy has therefore been a major focus of DM research. However, changes in gene expression, protein translation and micro-RNA metabolism may also contribute to disease pathology. The exciting finding of bidirectional transcription and non-conventional RNA translation of trinucleotide repeat sequences points to a new scenario, in which DM is not mediated by one single expanded RNA transcript, but involves multiple pathogenic elements and pathways. The study of the growing number of human diseases associated with toxic repeat-containing transcripts provides important insight into the understanding of the complex pathways of RNA toxicity. This review describes some of the recent advances in the understanding of the molecular mechanisms behind DM and other RNA-dominant disorders.
Collapse
Affiliation(s)
- Géraldine Sicot
- INSERM U781, Université Paris Descartes, Hôpital Necker Enfants Malades, 156 rue de Vaugirard, Paris Cedex 15, France
| | | | | |
Collapse
|
26
|
Abstract
The development of effective therapies for neuromuscular disorders such as Duchenne muscular dystrophy (DMD) is hampered by considerable challenges: skeletal muscle is the most abundant tissue in the body, and many neuromuscular disorders are multisystemic conditions. However, despite these barriers there has recently been substantial progress in the search for novel treatments. In particular, the use of antisense oligonucleotides, which are designed to target RNA and modulate pre-mRNA splicing to restore functional protein isoforms or directly inhibit the toxic effects of pathogenic RNAs, offers great promise and these approaches are now being tested in the clinic. Here, we review recent advances in the development of such antisense oligonucleotides and other promising novel approaches, including the induction of readthrough nonsense mutations.
Collapse
Affiliation(s)
- Francesco Muntoni
- UCL Institute of Child Health and Great Ormond Street Hospital, 30 Guildford Street, London WC1N 1EH, UK.
| | | |
Collapse
|
27
|
Udd B, Meola G, Krahe R, Wansink D, Bassez G, Kress W, Schoser B, Moxley R. Myotonic dystrophy type 2 (DM2) and related disorders. Neuromuscul Disord 2011; 21:443-50. [DOI: 10.1016/j.nmd.2011.03.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 03/31/2011] [Indexed: 01/18/2023]
|
28
|
Abnormal actomyosin assembly in proliferating and differentiating myoblasts upon expression of a cytosolic DMPK isoform. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:867-77. [PMID: 21295081 DOI: 10.1016/j.bbamcr.2011.01.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 01/05/2011] [Accepted: 01/24/2011] [Indexed: 01/17/2023]
Abstract
DMPK, the product of the mutated gene in myotonic dystrophy type 1, belongs to the subfamily of Rho-associated serine-threonine protein kinases, whose members play a role in actin-based cell morphodynamics. Not much is known about the physiological role of differentially localized individual DMPK splice isoforms. We report here that prominent stellar-shaped stress fibers are formed during early and late steps of differentiation in DMPK-deficient myoblast-myotubes upon complementation with the short cytosolic DMPK E isoform. Expression of DMPK E led to an increased phosphorylation status of MLC2. We found no such effects with vectors that encode a mutant DMPK E which was rendered enzymatically inactive or any of the long C-terminally anchored DMPK isoforms. Presence of stellar structures appears associated with changes in cell shape and motility and a delay in myogenesis. Our data strongly suggest that cytosolic DMPK participates in remodeling of the actomyosin cytoskeleton in developing skeletal muscle cells. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
|
29
|
Magaña JJ, Cisneros B. Perspectives on gene therapy in myotonic dystrophy type 1. J Neurosci Res 2010; 89:275-85. [DOI: 10.1002/jnr.22551] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 09/29/2010] [Accepted: 10/14/2010] [Indexed: 11/08/2022]
|
30
|
Cunningham CL, Martínez Cerdeño V, Navarro Porras E, Prakash AN, Angelastro JM, Willemsen R, Hagerman PJ, Pessah IN, Berman RF, Noctor SC. Premutation CGG-repeat expansion of the Fmr1 gene impairs mouse neocortical development. Hum Mol Genet 2010; 20:64-79. [PMID: 20935171 DOI: 10.1093/hmg/ddq432] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late adult-onset neurodegenerative disorder caused by a premutation CGG-trinucleotide repeat expansion (55-200 CGG repeats) within the 5'-untranslated region of the FMR1 gene. Although FXTAS generally affects premutation carriers over 50 years of age, cognitive and psychological symptoms can appear in carriers during childhood, suggesting that the FMR1 premutation affects brain function early in life. Recent work with cultured hippocampal neurons from a premutation (Fmr1 CGG knock-in) mouse model revealed impaired development of early postnatal neurons, consistent with the developmental clinical involvement of premutation carriers. In the current work, we show that the presence of premutation CGG-repeat expansions in the mouse Fmr1 gene alters embryonic neocortical development. Specifically, embryonic premutation mice display migration defects in the neocortex and altered expression of neuronal lineage markers. The current data demonstrate that premutation alleles of the Fmr1 gene are associated with defects in developmental programs operating during prenatal stages of brain formation and provide further evidence that the FMR1 premutation has a neurodevelopmental component.
Collapse
Affiliation(s)
- Christopher L Cunningham
- Department of Pathology, Institute for Pediatric Regenerative Medicine--Shriners Hospital, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|