1
|
Kelley CP, Haerle MC, Wang ET. Negative autoregulation mitigates collateral RNase activity of repeat-targeting CRISPR-Cas13d in mammalian cells. Cell Rep 2022; 40:111226. [PMID: 35977479 PMCID: PMC9809062 DOI: 10.1016/j.celrep.2022.111226] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/29/2022] [Accepted: 07/26/2022] [Indexed: 01/05/2023] Open
Abstract
CRISPR-Cas13 RNA endonucleases show promise for programmable RNA knockdown. However, sequence-specific binding of Cas13 unleashes non-specific bystander RNA cleavage, or collateral activity, raising concerns for experiments and therapeutic applications. Although robust in cell-free and bacterial environments, collateral activity in mammalian cells remains disputed. We investigate Cas13d collateral activity in a therapeutic context for myotonic dystrophy type 1, caused by a transcribed CTG repeat expansion. We find that, when targeting CUGn RNA in mammalian cells, Cas13d depletes endogenous and transgenic RNAs, interferes with critical cellular processes, and activates stress response and apoptosis. Collateral effects also occur when targeting abundant endogenous transcripts. To minimize collateral activity for repeat-targeting approaches, we introduce GENO, an adeno-associated virus-compatible strategy that leverages guide RNA processing to control Cas13d expression. We argue that thorough assessment of collateral activity is necessary when applying Cas13 in mammalian cells and that GENO illustrates advantages of compact regulatory systems for Cas-based gene therapies.
Collapse
Affiliation(s)
- Chase P Kelley
- Department of Molecular Genetics & Microbiology, Center for NeuroGenetics, Genetics Institute, University of Florida, Gainesville, FL 32608, USA; Genetics and Genomics Graduate Program, University of Florida, Gainesville, FL 32608, USA
| | - Maja C Haerle
- Department of Molecular Genetics & Microbiology, Center for NeuroGenetics, Genetics Institute, University of Florida, Gainesville, FL 32608, USA
| | - Eric T Wang
- Department of Molecular Genetics & Microbiology, Center for NeuroGenetics, Genetics Institute, University of Florida, Gainesville, FL 32608, USA; Myology Institute, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
2
|
Soltanzadeh P. Myotonic Dystrophies: A Genetic Overview. Genes (Basel) 2022; 13:367. [PMID: 35205411 PMCID: PMC8872148 DOI: 10.3390/genes13020367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophies (DM) are the most common muscular dystrophies in adults, which can affect other non-skeletal muscle organs such as the heart, brain and gastrointestinal system. There are two genetically distinct types of myotonic dystrophy: myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2), both dominantly inherited with significant overlap in clinical manifestations. DM1 results from CTG repeat expansions in the 3'-untranslated region (3'UTR) of the DMPK (dystrophia myotonica protein kinase) gene on chromosome 19, while DM2 is caused by CCTG repeat expansions in intron 1 of the CNBP (cellular nucleic acid-binding protein) gene on chromosome 3. Recent advances in genetics and molecular biology, especially in the field of RNA biology, have allowed better understanding of the potential pathomechanisms involved in DM. In this review article, core clinical features and genetics of DM are presented followed by a discussion on the current postulated pathomechanisms and therapeutic approaches used in DM, including the ones currently in human clinical trial phase.
Collapse
Affiliation(s)
- Payam Soltanzadeh
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Tanner MK, Tang Z, Thornton CA. Targeted splice sequencing reveals RNA toxicity and therapeutic response in myotonic dystrophy. Nucleic Acids Res 2021; 49:2240-2254. [PMID: 33503262 PMCID: PMC7913682 DOI: 10.1093/nar/gkab022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 01/03/2021] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Biomarker-driven trials hold promise for therapeutic development in chronic diseases, such as muscular dystrophy. Myotonic dystrophy type 1 (DM1) involves RNA toxicity, where transcripts containing expanded CUG-repeats (CUGexp) accumulate in nuclear foci and sequester splicing factors in the Muscleblind-like (Mbnl) family. Oligonucleotide therapies to mitigate RNA toxicity have emerged but reliable measures of target engagement are needed. Here we examined muscle transcriptomes in mouse models of DM1 and found that CUGexp expression or Mbnl gene deletion cause similar dysregulation of alternative splicing. We selected 35 dysregulated exons for further study by targeted RNA sequencing. Across a spectrum of mouse models, the individual splice events and a composite index derived from all events showed a graded response to decrements of Mbnl or increments of CUGexp. Antisense oligonucleotides caused prompt reduction of CUGexp RNA and parallel correction of the splicing index, followed by subsequent elimination of myotonia. These results suggest that targeted splice sequencing may provide a sensitive and reliable way to assess therapeutic impact in DM1.
Collapse
Affiliation(s)
- Matthew K Tanner
- Medical Scientist Training Program, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Zhenzhi Tang
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Charles A Thornton
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
4
|
Weskamp K, Olwin BB, Parker R. Post-Transcriptional Regulation in Skeletal Muscle Development, Repair, and Disease. Trends Mol Med 2020; 27:469-481. [PMID: 33384234 DOI: 10.1016/j.molmed.2020.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022]
Abstract
Skeletal muscle formation is a complex process that requires tight spatiotemporal control of key myogenic factors. Emerging evidence suggests that RNA processing is crucial for the regulation of these factors, and that multiple post-transcriptional regulatory pathways work dependently and independently of one another to enable precise control of transcripts throughout muscle development and repair. Moreover, disruption of these pathways is implicated in neuromuscular disease, and the recent development of RNA-mediated therapies shows enormous promise in the treatment of these disorders. We discuss the overlapping post-transcriptional regulatory pathways that mediate muscle development, how these pathways are disrupted in neuromuscular disorders, and advances in RNA-mediated therapies that present a novel approach to the treatment of these diseases.
Collapse
Affiliation(s)
- Kaitlin Weskamp
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA.
| | - Bradley B Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Roy Parker
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
5
|
Castro AF, Loureiro JR, Bessa J, Silveira I. Antisense Transcription across Nucleotide Repeat Expansions in Neurodegenerative and Neuromuscular Diseases: Progress and Mysteries. Genes (Basel) 2020; 11:E1418. [PMID: 33261024 PMCID: PMC7760973 DOI: 10.3390/genes11121418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Unstable repeat expansions and insertions cause more than 30 neurodegenerative and neuromuscular diseases. Remarkably, bidirectional transcription of repeat expansions has been identified in at least 14 of these diseases. More remarkably, a growing number of studies has been showing that both sense and antisense repeat RNAs are able to dysregulate important cellular pathways, contributing together to the observed clinical phenotype. Notably, antisense repeat RNAs from spinocerebellar ataxia type 7, myotonic dystrophy type 1, Huntington's disease and frontotemporal dementia/amyotrophic lateral sclerosis associated genes have been implicated in transcriptional regulation of sense gene expression, acting either at a transcriptional or posttranscriptional level. The recent evidence that antisense repeat RNAs could modulate gene expression broadens our understanding of the pathogenic pathways and adds more complexity to the development of therapeutic strategies for these disorders. In this review, we cover the amazing progress made in the understanding of the pathogenic mechanisms associated with repeat expansion neurodegenerative and neuromuscular diseases with a focus on the impact of antisense repeat transcription in the development of efficient therapies.
Collapse
Affiliation(s)
- Ana F. Castro
- Genetics of Cognitive Dysfunction Laboratory, i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.F.C.); (J.R.L.)
- IBMC-Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS, Universidade do Porto, 4050-313 Porto, Portugal
| | - Joana R. Loureiro
- Genetics of Cognitive Dysfunction Laboratory, i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.F.C.); (J.R.L.)
- IBMC-Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal;
| | - José Bessa
- IBMC-Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal;
- Vertebrate Development and Regeneration Laboratory, i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Isabel Silveira
- Genetics of Cognitive Dysfunction Laboratory, i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.F.C.); (J.R.L.)
- IBMC-Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal;
| |
Collapse
|
6
|
Navabpour S, Kwapis JL, Jarome TJ. A neuroscientist's guide to transgenic mice and other genetic tools. Neurosci Biobehav Rev 2020; 108:732-748. [PMID: 31843544 PMCID: PMC8049509 DOI: 10.1016/j.neubiorev.2019.12.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
The past decade has produced an explosion in the number and variety of genetic tools available to neuroscientists, resulting in an unprecedented ability to precisely manipulate the genome and epigenome in behaving animals. However, no single resource exists that describes all of the tools available to neuroscientists. Here, we review the genetic, transgenic, and viral techniques that are currently available to probe the complex relationship between genes and cognition. Topics covered include types of traditional transgenic mouse models (knockout, knock-in, reporter lines), inducible systems (Cre-loxP, Tet-On, Tet-Off) and cell- and circuit-specific systems (TetTag, TRAP, DIO-DREADD). Additionally, we provide details on virus-mediated and siRNA/shRNA approaches, as well as a comprehensive discussion of the myriad manipulations that can be made using the CRISPR-Cas9 system, including single base pair editing and spatially- and temporally-regulated gene-specific transcriptional control. Collectively, this review will serve as a guide to assist neuroscientists in identifying and choosing the appropriate genetic tools available to study the complex relationship between the brain and behavior.
Collapse
Affiliation(s)
- Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, College Park, PA, USA; Center for the Molecular Investigation of Neurological Disorders (CMIND), Pennsylvania State University, College Park, PA, USA.
| | - Timothy J Jarome
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
7
|
A CTG repeat-selective chemical screen identifies microtubule inhibitors as selective modulators of toxic CUG RNA levels. Proc Natl Acad Sci U S A 2019; 116:20991-21000. [PMID: 31570586 DOI: 10.1073/pnas.1901893116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A CTG repeat expansion in the DMPK gene is the causative mutation of myotonic dystrophy type 1 (DM1). Transcription of the expanded CTG repeat produces toxic gain-of-function CUG RNA, leading to disease symptoms. A screening platform that targets production or stability of the toxic CUG RNA in a selective manner has the potential to provide new biological and therapeutic insights. A DM1 HeLa cell model was generated that stably expresses a toxic r(CUG)480 and an analogous r(CUG)0 control from DMPK and was used to measure the ratio-metric level of r(CUG)480 versus r(CUG)0. This DM1 HeLa model recapitulates pathogenic hallmarks of DM1, including CUG ribonuclear foci and missplicing of pre-mRNA targets of the muscleblind (MBNL) alternative splicing factors. Repeat-selective screening using this cell line led to the unexpected identification of multiple microtubule inhibitors as hits that selectively reduce r(CUG)480 levels and partially rescue MBNL-dependent missplicing. These results were validated by using the Food and Drug Administration-approved clinical microtubule inhibitor colchicine in DM1 mouse and primary patient cell models. The mechanism of action was found to involve selective reduced transcription of the CTG expansion that we hypothesize to involve the LINC (linker of nucleoskeleton and cytoskeleton) complex. The unanticipated identification of microtubule inhibitors as selective modulators of toxic CUG RNA opens research directions for this form of muscular dystrophy and may shed light on the biology of CTG repeat expansion and inform therapeutic avenues. This approach has the potential to identify modulators of expanded repeat-containing gene expression for over 30 microsatellite expansion disorders.
Collapse
|
8
|
Turunen TA, Roberts TC, Laitinen P, Väänänen MA, Korhonen P, Malm T, Ylä-Herttuala S, Turunen MP. Changes in nuclear and cytoplasmic microRNA distribution in response to hypoxic stress. Sci Rep 2019; 9:10332. [PMID: 31316122 PMCID: PMC6637125 DOI: 10.1038/s41598-019-46841-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/05/2019] [Indexed: 02/08/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that have well-characterized roles in cytoplasmic gene regulation, where they act by binding to mRNA transcripts and inhibiting their translation (i.e. post-transcriptional gene silencing, PTGS). However, miRNAs have also been implicated in transcriptional gene regulation and alternative splicing, events that are restricted to the cell nucleus. Here we performed nuclear-cytoplasmic fractionation in a mouse endothelial cell line and characterized the localization of miRNAs in response to hypoxia using small RNA sequencing. A highly diverse population of abundant miRNA species was detected in the nucleus, of which the majority (56%) was found to be preferentially localized in one compartment or the other. Induction of hypoxia resulted in changes in miRNA levels in both nuclear and cytoplasmic compartments, with the majority of changes being restricted to one location and not the other. Notably, the classical hypoxamiR (miR-210-3p) was highly up-regulated in the nuclear compartment after hypoxic stimulus. These findings reveal a previously unappreciated level of molecular complexity in the physiological response occurring in ischemic tissue. Furthermore, widespread differential miRNA expression in the nucleus strongly suggests that these small RNAs are likely to perform extensive nuclear regulatory functions in the general case.
Collapse
Affiliation(s)
- Tiia A Turunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland
| | - Thomas C Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.,Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pia Laitinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland
| | - Mari-Anna Väänänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland.,Heart Center and Gene Therapy Unit, Kuopio University Hospital, PO Box 100, 70029 KUH, Kuopio, Finland
| | - Mikko P Turunen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Yliopistonranta 1E, 70210, Kuopio, Finland.
| |
Collapse
|
9
|
Abstract
Myotonic dystrophy is an autosomal dominant muscular dystrophy not only associated with muscle weakness, atrophy, and myotonia but also prominent multisystem involvement. There are 2 similar, but distinct, forms of myotonic dystrophy; type 1 is caused by a CTG repeat expansion in the DMPK gene, and type 2 is caused by a CCTG repeat expansion in the CNBP gene. Type 1 is associated with distal limb, neck flexor, and bulbar weakness and results in different phenotypic subtypes with variable onset from congenital to very late-onset as well as variable signs and symptoms. The classically described adult-onset form is the most common. In contrast, myotonic dystrophy type 2 is adult-onset or late-onset, has proximal predominant muscle weakness, and generally has less severe multisystem involvement. In both forms of myotonic dystrophy, the best characterized disease mechanism is a RNA toxic gain-of-function during which RNA repeats form nuclear foci resulting in sequestration of RNA-binding proteins and, therefore, dysregulated splicing of premessenger RNA. There are currently no disease-modifying therapies, but clinical surveillance, preventative measures, and supportive treatments are used to reduce the impact of muscular impairment and other systemic involvement including cataracts, cardiac conduction abnormalities, fatigue, central nervous system dysfunction, respiratory weakness, dysphagia, and endocrine dysfunction. Exciting preclinical progress has been made in identifying a number of potential strategies including genome editing, small molecule therapeutics, and antisense oligonucleotide-based therapies to target the pathogenesis of type 1 and type 2 myotonic dystrophies at the DNA, RNA, or downstream target level.
Collapse
Affiliation(s)
- Samantha LoRusso
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, Columbus, OH, 43210, USA
| | - Benjamin Weiner
- The Ohio State University College of Medicine, The Ohio State University, 370 West 9th Avenue, Columbus, OH, 43210, USA
| | - W David Arnold
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
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: 8.4] [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
|
11
|
RNA-mediated therapies in myotonic dystrophy. Drug Discov Today 2018; 23:2013-2022. [PMID: 30086404 DOI: 10.1016/j.drudis.2018.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/29/2022]
Abstract
Myotonic dystrophy 1 (DM1) is a multisystemic neuromuscular disease caused by a dominantly inherited 'CTG' repeat expansion in the gene encoding DM Protein Kinase (DMPK). The repeats are transcribed into mRNA, which forms hairpins and binds with high affinity to the Muscleblind-like (MBNL) family of proteins, sequestering them from their normal function. The loss of function of MBNL proteins causes numerous downstream effects, primarily the appearance of nuclear foci, mis-splicing, and ultimately myotonia and other clinical symptoms. Antisense and other RNA-mediated technologies have been applied to target toxic-repeat mRNA transcripts to restore MBNL protein function in DM1 models, such as cells and mice, and in humans. This technique has had promising results in DM1 therapeutics by alleviating pathogenic phenotypes.
Collapse
|
12
|
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
|
13
|
Dial AG, Ng SY, Manta A, Ljubicic V. The Role of AMPK in Neuromuscular Biology and Disease. Trends Endocrinol Metab 2018; 29:300-312. [PMID: 29572064 DOI: 10.1016/j.tem.2018.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a primary regulator of cellular metabolism. Recent studies have revealed that AMPK also mediates the maintenance and plasticity of α-motoneurons, the neuromuscular junction, and skeletal muscle. Furthermore, AMPK stimulation by either genetic, pharmacological, or physiological approaches elicits beneficial phenotypic remodeling in neuromuscular disorders (NMDs). Here, we review the role of AMPK as a governor of neuromuscular biology, and present evidence for AMPK as an effective molecular target for therapeutic pursuit in the context of the most prevalent NMDs, including Duchenne muscular dystrophy, spinal muscular atrophy, and myotonic dystrophy type 1. This information may be useful for engineering AMPK-targeted pharmacological- or lifestyle-based strategies to treat disorders of the neuromuscular system.
Collapse
Affiliation(s)
- Athan G Dial
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Alexander Manta
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
14
|
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
|
15
|
Fagg WS, Liu N, Fair JH, Shiue L, Katzman S, Donohue JP, Ares M. Autogenous cross-regulation of Quaking mRNA processing and translation balances Quaking functions in splicing and translation. Genes Dev 2017; 31:1894-1909. [PMID: 29021242 PMCID: PMC5695090 DOI: 10.1101/gad.302059.117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/11/2017] [Indexed: 12/18/2022]
Abstract
Quaking protein isoforms arise from a single Quaking gene and bind the same RNA motif to regulate splicing, translation, decay, and localization of a large set of RNAs. However, the mechanisms by which Quaking expression is controlled to ensure that appropriate amounts of each isoform are available for such disparate gene expression processes are unknown. Here we explore how levels of two isoforms, nuclear Quaking-5 (Qk5) and cytoplasmic Qk6, are regulated in mouse myoblasts. We found that Qk5 and Qk6 proteins have distinct functions in splicing and translation, respectively, enforced through differential subcellular localization. We show that Qk5 and Qk6 regulate distinct target mRNAs in the cell and act in distinct ways on their own and each other's transcripts to create a network of autoregulatory and cross-regulatory feedback controls. Morpholino-mediated inhibition of Qk translation confirms that Qk5 controls Qk RNA levels by promoting accumulation and alternative splicing of Qk RNA, whereas Qk6 promotes its own translation while repressing Qk5. This Qk isoform cross-regulatory network responds to additional cell type and developmental controls to generate a spectrum of Qk5/Qk6 ratios, where they likely contribute to the wide range of functions of Quaking in development and cancer.
Collapse
Affiliation(s)
- W Samuel Fagg
- Sinsheimer Laboratories, Department of Molecular, Cell, and Developmental Biology, Center for Molecular Biology of RNA, University of California at Santa Cruz. Santa Cruz, California 95064, USA.,Department of Surgery, Transplant Division, Shriners Hospital for Children, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Naiyou Liu
- Department of Surgery, Transplant Division, Shriners Hospital for Children, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Jeffrey Haskell Fair
- Department of Surgery, Transplant Division, Shriners Hospital for Children, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Lily Shiue
- Sinsheimer Laboratories, Department of Molecular, Cell, and Developmental Biology, Center for Molecular Biology of RNA, University of California at Santa Cruz. Santa Cruz, California 95064, USA
| | - Sol Katzman
- Sinsheimer Laboratories, Department of Molecular, Cell, and Developmental Biology, Center for Molecular Biology of RNA, University of California at Santa Cruz. Santa Cruz, California 95064, USA
| | - John Paul Donohue
- Sinsheimer Laboratories, Department of Molecular, Cell, and Developmental Biology, Center for Molecular Biology of RNA, University of California at Santa Cruz. Santa Cruz, California 95064, USA
| | - Manuel Ares
- Sinsheimer Laboratories, Department of Molecular, Cell, and Developmental Biology, Center for Molecular Biology of RNA, University of California at Santa Cruz. Santa Cruz, California 95064, USA
| |
Collapse
|
16
|
Thornton CA, Wang E, Carrell EM. Myotonic dystrophy: approach to therapy. Curr Opin Genet Dev 2017; 44:135-140. [PMID: 28376341 PMCID: PMC5447481 DOI: 10.1016/j.gde.2017.03.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/25/2017] [Accepted: 03/13/2017] [Indexed: 01/16/2023]
Abstract
Myotonic dystrophy (DM) is a dominantly-inherited genetic disorder affecting skeletal muscle, heart, brain, and other organs. DM type 1 is caused by expansion of a CTG triplet repeat in DMPK, whereas DM type 2 is caused by expansion of a CCTG tetramer repeat in CNBP. In both cases the DM mutations lead to expression of dominant-acting RNAs. Studies of RNA toxicity have now revealed novel mechanisms and new therapeutic targets. Preclinical data have suggested that RNA dominance is responsive to therapeutic intervention and that DM therapy can be approached at several different levels. Here we review recent efforts to alleviate RNA toxicity in DM.
Collapse
Affiliation(s)
- Charles A Thornton
- Department of Neurology, University of Rochester, Rochester 14642, NY, United States.
| | - Eric Wang
- Department of Molecular Genetics & Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, FL, United States
| | - Ellie M Carrell
- Department of Neurology, University of Rochester, Rochester 14642, NY, United States
| |
Collapse
|
17
|
Jauvin D, Chrétien J, Pandey SK, Martineau L, Revillod L, Bassez G, Lachon A, MacLeod AR, Gourdon G, Wheeler TM, Thornton CA, Bennett CF, Puymirat J. Targeting DMPK with Antisense Oligonucleotide Improves Muscle Strength in Myotonic Dystrophy Type 1 Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624222 PMCID: PMC5453865 DOI: 10.1016/j.omtn.2017.05.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Myotonic dystrophy type 1 (DM1), a dominant hereditary muscular dystrophy, is caused by an abnormal expansion of a (CTG)n trinucleotide repeat in the 3′ UTR of the human dystrophia myotonica protein kinase (DMPK) gene. As a consequence, mutant transcripts containing expanded CUG repeats are retained in nuclear foci and alter the function of splicing regulatory factors members of the MBNL and CELF families, resulting in alternative splicing misregulation of specific transcripts in affected DM1 tissues. In the present study, we treated DMSXL mice systemically with a 2′-4′-constrained, ethyl-modified (ISIS 486178) antisense oligonucleotide (ASO) targeted to the 3′ UTR of the DMPK gene, which led to a 70% reduction in CUGexp RNA abundance and foci in different skeletal muscles and a 30% reduction in the heart. Furthermore, treatment with ISIS 486178 ASO improved body weight, muscle strength, and muscle histology, whereas no overt toxicity was detected. This is evidence that the reduction of CUGexp RNA improves muscle strength in DM1, suggesting that muscle weakness in DM1 patients may be improved following elimination of toxic RNAs.
Collapse
Affiliation(s)
- Dominic Jauvin
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Jessina Chrétien
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Sanjay K Pandey
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA; Triangulum Biopharma, San Diego, CA 92121, USA
| | - Laurie Martineau
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada
| | - Lucille Revillod
- INSERM U955, Neuromuscular Reference Center, Henri-Mondor Hospital, Créteil 94000, France
| | - Guillaume Bassez
- INSERM U955, Neuromuscular Reference Center, Henri-Mondor Hospital, Créteil 94000, France
| | - Aline Lachon
- INSERM U781, Imagine Institute, Paris 75015, France
| | | | | | | | | | | | - Jack Puymirat
- Laval University Experimental Organogenesis Center/LOEX, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada; Department of Neurological Sciences CHU de Québec-Laval University, Enfant-Jésus Hospital, Québec, QC G1J 1Z4, Canada.
| |
Collapse
|
18
|
Abstract
Encapsulation of proteins in nanoparticles (NPs) can greatly improve the properties of proteins such as their stability against denaturation and degradation by proteases, and branches out the applications of natural proteins from their intrinsic localizations and functions in living organisms for biomedical and industrial applications. We recently developed several methods to armor proteins in NPs with sizes from nanometers up to >100nm, batch by batch or one by one, covalently or noncovalently, for a wide range of applications from biocatalysis to bioimaging and drug delivery. In this chapter, we provide detailed protocols on these methods. Key steps of specific protocols are explained with particular examples to help other laboratories to adopt and modify these methods for their own purposes. The advantages and disadvantages of each method are summarized, and guidelines for choosing the right method for a given application, as well as the current challenges and future directions of this field, are discussed.
Collapse
Affiliation(s)
- Yi Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai, China.
| |
Collapse
|
19
|
Tripathi BK, Surabhi S, Bhaskar PK, Mukherjee A, Mutsuddi M. The RNA binding KH domain of Spoonbill depletes pathogenic non-coding spinocerebellar ataxia 8 transcripts and suppresses neurodegeneration in Drosophila. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1732-41. [PMID: 27302466 DOI: 10.1016/j.bbadis.2016.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 06/06/2016] [Accepted: 06/09/2016] [Indexed: 12/26/2022]
Abstract
Spinocerebellar ataxia 8 (SCA8) pathogenesis is a resultant of gain-of-function machinery that primarily results at the RNA level. It has been reported that expanded non-coding CTG trinucleotide repeat in the ATXN8OS transcripts leads to SCA8 coupled neurodegeneration. Targeted depletion of pathogenic SCA8 transcripts is a viable therapeutic approach. In this report we have focused on the suppression of toxic RNA gain-of-function associated with SCA8. We report suppression of SCA8 associated neurodegeneration by KH RNA binding domain of Spoonbill. KH domain suppresses pathogenic SCA8 associated phenotype in adult flies. Ectopic expression of KH domain leads to massive reduction in the number and size of SCA8 RNA foci. We show that Spoonbill interacts with toxic SCA8 transcripts via its KH domain and promotes its depletion. Till date, no attempts have been made for therapeutic intervention of SCA8 pathogenesis. Further characterization of Spoonbill KH domain may aid us in designing peptide based therapeutics for SCA8 associated neurodegeneration.
Collapse
Affiliation(s)
- Bipin K Tripathi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Satya Surabhi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Pradeep K Bhaskar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| |
Collapse
|
20
|
Koscianska E, Witkos TM, Kozlowska E, Wojciechowska M, Krzyzosiak WJ. Cooperation meets competition in microRNA-mediated DMPK transcript regulation. Nucleic Acids Res 2015; 43:9500-18. [PMID: 26304544 PMCID: PMC4627076 DOI: 10.1093/nar/gkv849] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/10/2015] [Indexed: 02/07/2023] Open
Abstract
The fundamental role of microRNAs (miRNAs) in the regulation of gene expression has been well-established, but many miRNA-driven regulatory mechanisms remain elusive. In the present study, we demonstrate that miRNAs regulate the expression of DMPK, the gene mutated in myotonic dystrophy type 1 (DM1), and we provide insight regarding the concerted effect of the miRNAs on the DMPK target. Specifically, we examined the binding of several miRNAs to the DMPK 3′ UTR using luciferase assays. We validated the interactions between the DMPK transcript and the conserved miR-206 and miR-148a. We suggest a possible cooperativity between these two miRNAs and discuss gene targeting by miRNA pairs that vary in distance between their binding sites and expression profiles. In the same luciferase reporter system, we showed miR-15b/16 binding to the non-conserved CUG repeat tract present in the DMPK transcript and that the CUG-repeat-binding miRNAs might also act cooperatively. Moreover, we detected miR-16 in cytoplasmic foci formed by exogenously expressed RNAs with expanded CUG repeats. Therefore, we propose that the expanded CUGs may serve as a target for concerted regulation by miRNAs and may also act as molecular sponges for natural miRNAs with CAG repeats in their seed regions, thereby affecting their physiological functions.
Collapse
Affiliation(s)
- Edyta Koscianska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Tomasz M Witkos
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Emilia Kozlowska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Marzena Wojciechowska
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Wlodzimierz J Krzyzosiak
- Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
21
|
Liu Y, Song ZM, Deng X, Cui Y, Yang YF, Han K, Jin R, Wang H, Liu Y, Cao A. Chitosan-coated red fluorescent protein nanoparticle as a potential dual-functional siRNA carrier. Nanomedicine (Lond) 2015; 10:2005-16. [PMID: 26135851 DOI: 10.2217/nnm.15.56] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS Developing safe and efficient nano vectors is critical for the success of siRNA therapy. MATERIALS & METHODS By encapsulating red fluorescent protein (RFP) with chitosan (CS), a dual-functional siRNA delivery nano vector, RFP@CS, has been synthesized. RESULTS RFP@CS has an optimum size of 7-23 nm for siRNA delivery; and the fluorescence of RFP, protected by CS coating, provides an excellent probe to track the delivery of siRNA. RFP@CS delivers siRNA efficiently into cells and the targeted gene could be completely silenced even after 48 h. No cytotoxicity or acute toxicity in mice was observed. CONCLUSION The high transfection efficacy and safety demonstrate RFP@CS is a promising nano vector for the gene therapy.
Collapse
Affiliation(s)
- Ying Liu
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Zheng-Mei Song
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Xiaoyong Deng
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Yangdong Cui
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Yi-Fan Yang
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Kai Han
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Rong Jin
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Haifang Wang
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| | - Yuanfang Liu
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
- Beijing National Laboratory for Molecular Sciences, College of Chemistry & Molecular Engineering, Peking University, Beijing 100871, China
| | - Aoneng Cao
- Institute of Nanochemistry & Nanobiology, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
22
|
Bisset DR, Stepniak-Konieczna EA, Zavaljevski M, Wei J, Carter GT, Weiss MD, Chamberlain JR. Therapeutic impact of systemic AAV-mediated RNA interference in a mouse model of myotonic dystrophy. Hum Mol Genet 2015; 24:4971-83. [PMID: 26082468 DOI: 10.1093/hmg/ddv219] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/08/2015] [Indexed: 01/22/2023] Open
Abstract
RNA interference (RNAi) offers a promising therapeutic approach for dominant genetic disorders that involve gain-of-function mechanisms. One candidate disease for RNAi therapy application is myotonic dystrophy type 1 (DM1), which results from toxicity of a mutant mRNA. DM1 is caused by expansion of a CTG repeat in the 3' UTR of the DMPK gene. The expression of DMPK mRNA containing an expanded CUG repeat (CUG(exp)) leads to defects in RNA biogenesis and turnover. We designed miRNA-based RNAi hairpins to target the CUG(exp) mRNA in the human α-skeletal muscle actin long-repeat (HSA(LR)) mouse model of DM1. RNAi expression cassettes were delivered to HSA(LR) mice using recombinant adeno-associated viral (rAAV) vectors injected intravenously as a route to systemic gene therapy. Vector delivery significantly reduced disease pathology in muscles of the HSA(LR) mice, including a reduction in the CUG(exp) mRNA, a reduction in myotonic discharges, a shift toward adult pre-mRNA splicing patterns, reduced myofiber hypertrophy and a decrease in myonuclear foci containing the CUG(exp) mRNA. Significant reversal of hallmarks of DM1 in the rAAV RNAi-treated HSA(LR) mice indicate that defects characteristic of DM1 can be mitigated with a systemic RNAi approach targeting the nuclei of terminally differentiated myofibers. Efficient rAAV-mediated delivery of RNAi has the potential to provide a long-term therapy for DM1 and other dominant muscular dystrophies.
Collapse
Affiliation(s)
| | | | | | - Jessica Wei
- Division of Medical Genetics, Department of Medicine
| | | | - Michael D Weiss
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
23
|
Rigo F, Seth PP, Bennett CF. Antisense oligonucleotide-based therapies for diseases caused by pre-mRNA processing defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 825:303-52. [PMID: 25201110 DOI: 10.1007/978-1-4939-1221-6_9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Before a messenger RNA (mRNA) is translated into a protein in the cytoplasm, its pre-mRNA precursor is extensively processed through capping, splicing and polyadenylation in the nucleus. Defects in the processing of pre-mRNAs due to mutations in RNA sequences often cause disease. Traditional small molecules or protein-based therapeutics are not well suited for correcting processing defects by targeting RNA. However, antisense oligonucleotides (ASOs) designed to bind RNA by Watson-Crick base pairing can target most RNA transcripts and have emerged as the ideal therapeutic agents for diseases that are caused by pre-mRNA processing defects. Here we review the diverse ASO-based mechanisms that can be exploited to modulate the expression of RNA. We also discuss how advancements in medicinal chemistry and a deeper understanding of the pharmacokinetic and toxicological properties of ASOs have enabled their use as therapeutic agents. We end by describing how ASOs have been used successfully to treat various pre-mRNA processing diseases in animal models.
Collapse
Affiliation(s)
- Frank Rigo
- Isis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, USA,
| | | | | |
Collapse
|
24
|
Nascimento AV, Singh A, Bousbaa H, Ferreira D, Sarmento B, Amiji MM. Mad2 checkpoint gene silencing using epidermal growth factor receptor-targeted chitosan nanoparticles in non-small cell lung cancer model. Mol Pharm 2014; 11:3515-27. [PMID: 25256346 PMCID: PMC4186685 DOI: 10.1021/mp5002894] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
RNA
interference has emerged as a powerful strategy in cancer therapy
because it allows silencing of specific genes associated with tumor
progression and resistance. Mad2 is an essential mitotic checkpoint
component required for accurate chromosome segregation during mitosis,
and its complete abolition leads to cell death. We have developed
an epidermal growth factor receptor (EGFR)-targeted chitosan system
for silencing the Mad2 gene as a strategy to efficiently
induce cell death in EGFR overexpressing human A549 non-small cell
lung cancer cells. Control and EGFR-targeted chitosan nanoparticles
loaded with small interfering RNAs (siRNAs) against Mad2 were formulated
and characterized for size, charge, morphology, and encapsulation
efficiency. Qualitative and quantitative intracellular uptake studies
by confocal imaging and flow cytometry, respectively, showed time-dependent
enhanced and selective intracellular internalization of EGFR-targeted
nanoparticles compared to nontargeted system. Targeted nanoparticles
showed nearly complete depletion of Mad2 expression in A549 cells
contrasting with the partial depletion in the nontargeted system.
Accordingly, Mad2-silencing-induced apoptotic cell death was confirmed
by cytotoxicity assay and flow cytometry. Our results demonstrate
that EGFR-targeted chitosan loaded with Mad2 siRNAs
is a potent delivery system for selective killing of cancer cells.
Collapse
Affiliation(s)
- Ana Vanessa Nascimento
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, IINFACTS, Rua Central de Gandra 1317, 4585-116 Gandra PRD, Portugal
| | | | | | | | | | | |
Collapse
|
25
|
Roberts TC. The MicroRNA Biology of the Mammalian Nucleus. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e188. [PMID: 25137140 PMCID: PMC4221600 DOI: 10.1038/mtna.2014.40] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/08/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are a class of genome-encoded small RNAs that are primarily considered to be post-transcriptional negative regulators of gene expression acting in the cytoplasm. Over a decade of research has focused on this canonical paradigm of miRNA function, with many success stories. Indeed, miRNAs have been identified that act as master regulators of a myriad of cellular processes, and many miRNAs are promising therapeutic targets or disease biomarkers. However, it is becoming increasingly apparent that the canonical view of miRNA function is incomplete. Several lines of evidence now point to additional functions for miRNAs in the nucleus of the mammalian cell. The majority of cellular miRNAs are present in both the nucleus and the cytoplasm, and certain miRNAs show specific nuclear enrichment. Additionally, some miRNAs colocalize with sub-nuclear structures such as the nucleolus and chromatin. Multiple components of the miRNA processing machinery are present in the nuclear compartment and are shuttled back and forth across the nuclear envelope. In the nucleus, miRNAs act to regulate the stability of nuclear transcripts, induce epigenetic alterations that either silence or activate transcription at specific gene promoters, and modulate cotranscriptional alternative splicing events. Nuclear miRNA-directed gene regulation constitutes a departure from the prevailing view of miRNA function and as such, warrants detailed further investigation.
Collapse
Affiliation(s)
- Thomas C Roberts
- 1] Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA [2] Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Braun S. Gene-based therapies of neuromuscular disorders: an update and the pivotal role of patient organizations in their discovery and implementation. J Gene Med 2014; 15:397-413. [PMID: 24123726 DOI: 10.1002/jgm.2747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 12/13/2022] Open
Abstract
This review updates the state-of-the art accomplishments of the multifaceted gene-based therapies, which include DNA or RNA as either therapeutic tools or targets for the treatment of neuromuscular diseases. It also provides insights into the key role that patient organizations have played in research and development; in particular, by addressing bottlenecks and generating boundary conditions that have contributed to scientific breakthroughs, and the effectiveness of innovation processes. Several gene therapy methods have reached the clinical stage and are now addressing both specific and classical issues related to this novel technology. Not ready yet for clinical application, genome editing is at its infancy. More rapidly progressing, RNA-based therapeutics, and especially exon skipping, exon inclusion and stop codon readthrough strategies, are about to move to the market. Most importantly, patients were at the forefront of this discovery process, from basic knowledge to innovation and translational research in a rapidly growing field of unmet medical needs. In recent years, Duchenne muscular dystrophy was the fertile ground for new therapeutic concepts that have been extended to other neuromuscular disorders, such as spinal muscular atrophy, myotonic dystrophies or fascioscapulohumeral dystrophy. In line with their longstanding policy, patient organizations will keep working in a proactive manner to bring together all stakeholders with a view to working out truly therapeutic solutions over a long-term perspective.
Collapse
|
27
|
Coonrod LA, Nakamori M, Wang W, Carrell S, Hilton CL, Bodner MJ, Siboni RB, Docter AG, Haley MM, Thornton CA, Berglund JA. Reducing levels of toxic RNA with small molecules. ACS Chem Biol 2013; 8:2528-37. [PMID: 24028068 DOI: 10.1021/cb400431f] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Myotonic dystrophy (DM) is one of the most common forms of muscular dystrophy. DM is an autosomal dominant disease caused by a toxic gain of function RNA. The toxic RNA is produced from expanded noncoding CTG/CCTG repeats, and these CUG/CCUG repeats sequester the Muscleblind-like (MBNL) family of RNA binding proteins. The MBNL proteins are regulators of alternative splicing, and their sequestration has been linked with mis-splicing events in DM. A previously reported screen for small molecules found that pentamidine was able to improve splicing defects associated with DM. Biochemical experiments and cell and mouse model studies of the disease indicate that pentamidine and related compounds may work through binding the CTG*CAG repeat DNA to inhibit transcription. Analysis of a series of methylene linker analogues of pentamidine revealed that heptamidine reverses splicing defects and rescues myotonia in a DM1 mouse model.
Collapse
Affiliation(s)
| | - Masayuki Nakamori
- Department
of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Wenli Wang
- Department
of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Samuel Carrell
- Department
of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | | | | | | | | | | | - Charles A. Thornton
- Department
of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | | |
Collapse
|
28
|
Alvarez ML, Khosroheidari M, Eddy E, Kiefer J. Role of microRNA 1207-5P and its host gene, the long non-coding RNA Pvt1, as mediators of extracellular matrix accumulation in the kidney: implications for diabetic nephropathy. PLoS One 2013; 8:e77468. [PMID: 24204837 PMCID: PMC3808414 DOI: 10.1371/journal.pone.0077468] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/02/2013] [Indexed: 01/13/2023] Open
Abstract
Diabetic nephropathy is the most common cause of chronic kidney failure and end-stage renal disease in the Western World. One of the major characteristics of this disease is the excessive accumulation of extracellular matrix (ECM) in the kidney glomeruli. While both environmental and genetic determinants are recognized for their role in the development of diabetic nephropathy, epigenetic factors, such as DNA methylation, long non-coding RNAs, and microRNAs, have also recently been found to underlie some of the biological mechanisms, including ECM accumulation, leading to the disease. We previously found that a long non-coding RNA, the plasmacytoma variant translocation 1 (PVT1), increases plasminogen activator inhibitor 1 (PAI-1) and transforming growth factor beta 1 (TGF-β1) in mesangial cells, the two main contributors to ECM accumulation in the glomeruli under hyperglycemic conditions, as well as fibronectin 1 (FN1), a major ECM component. Here, we report that miR-1207-5p, a PVT1-derived microRNA, is abundantly expressed in kidney cells, and is upregulated by glucose and TGF-β1. We also found that like PVT1, miR-1207-5p increases expression of TGF-β1, PAI-1, and FN1 but in a manner that is independent of its host gene. In addition, regulation of miR-1207-5p expression by glucose and TGFβ1 is independent of PVT1. These results provide evidence supporting important roles for miR-1207-5p and its host gene in the complex pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- M. Lucrecia Alvarez
- Diabetes, Cardiovascular, and Metabolic Diseases Center, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
- * E-mail:
| | - Mahdieh Khosroheidari
- Diabetes, Cardiovascular, and Metabolic Diseases Center, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Elena Eddy
- Diabetes, Cardiovascular, and Metabolic Diseases Center, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jeff Kiefer
- Diabetes, Cardiovascular, and Metabolic Diseases Center, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| |
Collapse
|
29
|
Oana K, Oma Y, Suo S, Takahashi MP, Nishino I, Takeda S, Ishiura S. Manumycin A corrects aberrant splicing of Clcn1 in myotonic dystrophy type 1 (DM1) mice. Sci Rep 2013; 3:2142. [PMID: 23828222 PMCID: PMC3701899 DOI: 10.1038/srep02142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/17/2013] [Indexed: 01/12/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults and as yet no cure for DM1. Here, we report the potential of manumycin A for a novel DM1 therapeutic reagent. DM1 is caused by expansion of CTG repeat. Mutant transcripts containing expanded CUG repeats lead to aberrant regulation of alternative splicing. Myotonia (delayed muscle relaxation) is the most commonly observed symptom in DM1 patients and is caused by aberrant splicing of the skeletal muscle chloride channel (CLCN1) gene. Identification of small-molecule compounds that correct aberrant splicing in DM1 is attracting much attention as a way of improving understanding of the mechanism of DM1 pathology and improving treatment of DM1 patients. In this study, we generated a reporter screening system and searched for small-molecule compounds. We found that manumycin A corrects aberrant splicing of Clcn1 in cell and mouse models of DM1.
Collapse
Affiliation(s)
- Kosuke Oana
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Gao Z, Cooper TA. Antisense oligonucleotides: rising stars in eliminating RNA toxicity in myotonic dystrophy. Hum Gene Ther 2013; 24:499-507. [PMID: 23252746 DOI: 10.1089/hum.2012.212] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myotonic dystrophy (DM) is a dominantly inherited, multisystemic disease caused by expanded CTG (type 1, DM1) or CCTG (type 2, DM2) repeats in untranslated regions of the mutated genes. Pathogenesis results from expression of RNAs from the mutated alleles that are toxic because of the expanded CUG or CCUG repeats. Increased understanding of the repeat-containing RNA (C/CUG(exp) RNA)-induced toxicity has led to the development of multiple strategies targeting the toxic RNA. Among these approaches, antisense oligonucleotides (ASOs) have demonstrated high potency in reversing the RNA toxicity in both cultured DM1 cells and DM1 animal models, thus offering great promise for the potential treatment of DM1. ASO targeting approaches will also provide avenues for the treatment of other repeat RNA-mediated diseases.
Collapse
Affiliation(s)
- Zhihua Gao
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
31
|
Sobczak K, Wheeler TM, Wang W, Thornton CA. RNA interference targeting CUG repeats in a mouse model of myotonic dystrophy. Mol Ther 2012. [PMID: 23183533 DOI: 10.1038/mt.2012.222] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is an RNA dominant disease caused by expression of DM protein kinase (DMPK) transcripts that contain an expanded CUG repeat (CUG(exp)). The toxic mRNA localizes to nuclear foci and sequesters proteins involved in the regulation of alternative splicing, such as, muscleblind-like 1 (MBNL1). Here, we used synthetic short interfering RNAs (siRNAs) to target CUG repeats and test the concept that inhibiting the expression of CUG(exp) RNA can mitigate features of DM1 in transgenic mice. Intramuscular injection and electroporation of siRNA resulted in ~70-80% downregulation of CUG(exp) transcripts. A limited survey of endogenous mouse transcripts that contain nonexpanded CUG or CAG repeats showed that most were not affected, though Txlnb containing (CUG)(9) was significantly reduced. By this strategy, the number and intensity of CUG(exp) nuclear foci were reduced and splicing of MBNL1-dependent exons was improved. These data suggest that the expanded CUG repeats are a potential target for allele-selective RNA interference.
Collapse
Affiliation(s)
- Krzysztof Sobczak
- Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | | | |
Collapse
|
32
|
Zhang XX, Eden HS, Chen X. Peptides in cancer nanomedicine: drug carriers, targeting ligands and protease substrates. J Control Release 2012; 159:2-13. [PMID: 22056916 PMCID: PMC3288222 DOI: 10.1016/j.jconrel.2011.10.023] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/18/2011] [Indexed: 01/22/2023]
Abstract
Peptides are attracting increasing attention as therapeutic agents, as the technologies for peptide development and manufacture continue to mature. Concurrently, with booming research in nanotechnology for biomedical applications, peptides have been studied as an important class of components in nanomedicine, and they have been used either alone or in combination with nanomaterials of every reported composition. Peptides possess many advantages, such as smallness, ease of synthesis and modification, and good biocompatibility. Their functions in cancer nanomedicine, discussed in this review, include serving as drug carriers, as targeting ligands, and as protease-responsive substrates for drug delivery.
Collapse
Affiliation(s)
- Xiao-Xiang Zhang
- Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
33
|
miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J 2011; 30:4414-22. [PMID: 21964070 DOI: 10.1038/emboj.2011.359] [Citation(s) in RCA: 808] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 08/29/2011] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are ∼22 nt non-coding RNAs that typically bind to the 3' UTR of target mRNAs in the cytoplasm, resulting in mRNA destabilization and translational repression. Here, we report that miRNAs can also regulate gene expression by targeting non-coding antisense transcripts in human cells. Specifically, we show that miR-671 directs cleavage of a circular antisense transcript of the Cerebellar Degeneration-Related protein 1 (CDR1) locus in an Ago2-slicer-dependent manner. The resulting downregulation of circular antisense has a concomitant decrease in CDR1 mRNA levels, independently of heterochromatin formation. This study provides the first evidence for non-coding antisense transcripts as functional miRNA targets, and a novel regulatory mechanism involving a positive correlation between mRNA and antisense circular RNA levels.
Collapse
|
34
|
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
|
35
|
The miRNA pathway in neurological and skeletal muscle disease: implications for pathogenesis and therapy. J Mol Med (Berl) 2011; 89:1065-77. [PMID: 21751030 DOI: 10.1007/s00109-011-0781-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 06/14/2011] [Accepted: 06/14/2011] [Indexed: 01/10/2023]
Abstract
RNA interference (RNAi) represents a powerful post-transcriptional gene silencing network which fine-tunes gene expression in all eukaryotic cells. The endogenous triggers of RNAi, microRNAs (miRNAs), are proposed to regulate expression of up to a third of all protein-coding genes, and have been shown to have critical roles in developmental processes including in the central nervous system and skeletal muscle. Further, many have been reported to display differential expression in various disease states. Here we describe present understanding of the biogenesis and function of miRNAs, review current knowledge of miRNA abnormalities in both human neurological and skeletal muscle disease and discuss their potential as novel disease biomarkers. Finally, we highlight the many ways in which the miRNA pathway may be targeted for therapeutic benefit.
Collapse
|
36
|
Foff EP, Mahadevan MS. Therapeutics development in myotonic dystrophy type 1. Muscle Nerve 2011; 44:160-9. [PMID: 21607985 DOI: 10.1002/mus.22090] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2011] [Indexed: 01/06/2023]
Abstract
Myotonic dystrophy (DM1), the most common adult muscular dystrophy, is a multisystem, autosomal dominant genetic disorder caused by an expanded CTG repeat that leads to nuclear retention of a mutant RNA and subsequent RNA toxicity. Significant insights into the molecular mechanisms of RNA toxicity have led to the previously unforeseen possibility that treating DM1 is a viable prospect. In this review, we briefly present the clinical picture in DM1, and describe how the research in understanding the pathogenesis of RNA toxicity in DM1 has led to targeted approaches to therapeutic development at various steps in the pathogenesis of the disease. We discuss the promise and current limitations of each with an emphasis on RNA-based therapeutics and small molecules. We conclude with a discussion of the unmet need for clinical tools and outcome measures that are essential prerequisites to proceed in evaluating these potential therapies in clinical trials.
Collapse
Affiliation(s)
- Erin Pennock Foff
- Department of Neurology, University of Virginia, Charlottesville, Virginia, USA
| | | |
Collapse
|
37
|
Seinen E, Burgerhof JGM, Jansen RC, Sibon OCM. RNAi-induced off-target effects in Drosophila melanogaster: frequencies and solutions. Brief Funct Genomics 2011; 10:206-14. [PMID: 21596801 DOI: 10.1093/bfgp/elr017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Genes can be silenced with short-interfering RNA molecules (siRNA). siRNAs are widely used to identify gene functions and have high potential for therapeutic treatments. It is critical that the siRNA specifically targets the expression of the gene of interest but has no off-target effects on other genes. Although siRNAs were initially considered to be exclusively active on mature mRNAs in the cytoplasm, additional studies have shown that siRNAs are present in the nucleus as well, suggesting that pre-mRNA sequences containing introns and other untranslated regions can also be targeted. In this study, we investigated the extent to which off-targets may occur in Drosophila melanogaster by looking at mature mRNA sequences and pre-mature RNA sequences separately. First, an in silico approach revealed that, based on sequence similarity, numerous off-targets are predicted to occur in RNAi experiments. Second, existing microarray data were used to investigate a possible effect of the predicted off-targets based on analysis of in vitro data. We found that the occurrence of off-targets in both mature and pre-mature RNA sequences in RNAi experiments can be extensive and significant. Possibilities are discussed how to minimize off-target effects.
Collapse
Affiliation(s)
- Erwin Seinen
- Section of Radiation & Stress Cell Biology, Department of Cell Biology, University Medical Center Groningen, The Netherlands
| | | | | | | |
Collapse
|
38
|
Seyhan AA. RNAi: a potential new class of therapeutic for human genetic disease. Hum Genet 2011; 130:583-605. [PMID: 21537948 DOI: 10.1007/s00439-011-0995-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/17/2011] [Indexed: 12/19/2022]
Abstract
Dominant negative genetic disorders, in which a mutant allele of a gene causes disease in the presence of a second, normal copy, have been challenging since there is no cure and treatments are only to alleviate the symptoms. Current therapies involving pharmacological and biological drugs are not suitable to target mutant genes selectively due to structural indifference of the normal variant of their targets from the disease-causing mutant ones. In instances when the target contains single nucleotide polymorphism (SNP), whether it is an enzyme or structural or receptor protein are not ideal for treatment using conventional drugs due to their lack of selectivity. Therefore, there is a need to develop new approaches to accelerate targeting these previously inaccessible targets by classical therapeutics. Although there is a cooling trend by the pharmaceutical industry for the potential of RNA interference (RNAi), RNAi and other RNA targeting drugs (antisense, ribozyme, etc.) still hold their promise as the only drugs that provide an opportunity to target genes with SNP mutations found in dominant negative disorders, genes specific to pathogenic tumor cells, and genes that are critical for mediating the pathology of various other diseases. Because of its exquisite specificity and potency, RNAi has attracted a considerable interest as a new class of therapeutic for genetic diseases including amyotrophic lateral sclerosis, Huntington's disease (HD), Alzheimer's disease (AD), Parkinson's disease (PD), spinocerebellar ataxia, dominant muscular dystrophies, and cancer. In this review, progress and challenges in developing RNAi therapeutics for genetic diseases will be discussed.
Collapse
Affiliation(s)
- Attila A Seyhan
- Pfizer Inc., Translational Immunology, Inflammation and Immunology, 200 Cambridgepark Drive, Cambridge, MA 02140, USA.
| |
Collapse
|
39
|
Santoro F, Barlow DP. Developmental control of imprinted expression by macro non-coding RNAs. Semin Cell Dev Biol 2011; 22:328-35. [PMID: 21333747 DOI: 10.1016/j.semcdb.2011.02.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 02/11/2011] [Indexed: 01/22/2023]
Abstract
Genomic imprinting is a developmentally regulated epigenetic phenomenon. The majority of imprinted genes only show parent-of-origin specific expression in a subset of tissues or at defined developmental stages. In some cases, imprinted expression is controlled by an imprinted macro non-coding RNA (ncRNA) whose expression pattern and repressive activity does not necessarily correlate with that of the genes whose imprinted expression it controls. This suggests that developmentally regulated factors other than the macro ncRNA are involved in establishing or maintaining imprinted expression. Here, we review how macro ncRNAs control imprinted expression during development and differentiation and consider how this impacts on target choice in epigenetic therapy.
Collapse
Affiliation(s)
- Federica Santoro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Science, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria
| | | |
Collapse
|
40
|
Abstract
Myotonic dystrophies (dystrophia myotonica, or DM) are inherited disorders characterized by myotonia and progressive muscle degeneration, which are variably associated with a multisystemic phenotype. To date, two types of myotonic dystrophy, type 1 (DM1) and type 2 (DM2), are known to exist; both are autosomal dominant disorders caused by expansion of an untranslated short tandem repeat DNA sequence (CTG)(n) and (CCTG)(n), respectively. These expanded repeats in DM1 and DM2 show different patterns of repeat-size instability. Phenotypes of DM1 and DM2 are similar but there are some important differences, most conspicuously in the severity of the disease (including the presence or absence of the congenital form), muscles primarily affected (distal versus proximal), involved muscle fiber types (type 1 versus type 2 fibers), and some associated multisystemic phenotypes. The pathogenic mechanism of DM1 and DM2 is thought to be mediated by the mutant RNA transcripts containing expanded CUG and CCUG repeats. Strong evidence supports the hypothesis that sequestration of muscle-blind like (MBNL) proteins by these expanded repeats leads to misregulated splicing of many gene transcripts in corroboration with the raised level of CUG-binding protein 1. However, additional mechanisms, such as changes in the chromatin structure involving CTCN-binding site and gene expression dysregulations, are emerging. Although treatment of DM1 and DM2 is currently limited to supportive therapies, new therapeutic approaches based on pathogenic mechanisms may become feasible in the near future.
Collapse
Affiliation(s)
- Tetsuo Ashizawa
- Department of Neurology, McKnight Brain Institute, The University of Texas Medical Branch, Galveston, TX, USA.
| | | |
Collapse
|
41
|
Itoh K, Mitani M, Kawamoto K, Futamura N, Funakawa I, Jinnai K, Fushiki S. Neuropathology does not Correlate with Regional Differences in the Extent of Expansion of CTG Repeats in the Brain with Myotonic Dystrophy Type 1. Acta Histochem Cytochem 2010; 43:149-56. [PMID: 21245981 PMCID: PMC3015052 DOI: 10.1267/ahc.10019] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 11/01/2010] [Indexed: 11/22/2022] Open
Abstract
Myotonic dystrophy (DM1) is known to be an adult-onset muscular dystrophy caused by the expansion of CTG repeats within the 3' untranslated region of the dystrophin myotonin protein kinase (DMPK) gene. The clinical features of DM1 include CNS symptoms, such as cognitive impairment and personality changes, the pathogenesis of which remains to be elucidated. We hypothesized that the distribution of neuropathological changes might be correlated with the extent of the length of the CTG repeats in the DMPK genes in DM1 patients. We studied the neuropathological changes in the brains of subjects with DM1 and investigated the extent of somatic instability in terms of CTG repeat expansion in the different brain regions of the same individuals by Southern blot analysis. The neuropathological changes included état criblé in the cerebral deep white matter and neurofibrillary tangles immunoreactive for phosphorylated tau in the hippocampus and entorhinal cortex, both of which were compatible with the subcortical dementia in DM1 patients. However, the length of the CTG repeats did not correlate with the regional differences in the extent of neuropathological changes. Our data suggested that pathomechanisms of dementia in DM1 might be more multifactorial rather than a toxic gain-of-function due to mutant RNA.
Collapse
Affiliation(s)
- Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Maki Mitani
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital
| | - Kunihiko Kawamoto
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital
| | - Naonobu Futamura
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital
| | - Itaru Funakawa
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital
| | - Kenji Jinnai
- Department of Neurology, National Hospital Organization Hyogo-Chuo National Hospital
| | - Shinji Fushiki
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| |
Collapse
|
42
|
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: 1.3] [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]
|
43
|
XVIII Annual Congress of the European Society of Gene and Cell Therapy (ESGCT)October 22–25, 2010Milan, Italy. Hum Gene Ther 2010. [DOI: 10.1089/hum.2010.915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
44
|
Seinen E, Burgerhof JGM, Jansen RC, Sibon OCM. RNAi experiments in D. melanogaster: solutions to the overlooked problem of off-targets shared by independent dsRNAs. PLoS One 2010; 5. [PMID: 20957038 PMCID: PMC2948504 DOI: 10.1371/journal.pone.0013119] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 09/01/2010] [Indexed: 11/19/2022] Open
Abstract
Background RNAi technology is widely used to downregulate specific gene products. Investigating the phenotype induced by downregulation of gene products provides essential information about the function of the specific gene of interest. When RNAi is applied in Drosophila melanogaster or Caenorhabditis elegans, often large dsRNAs are used. One of the drawbacks of RNAi technology is that unwanted gene products with sequence similarity to the gene of interest can be down regulated too. To verify the outcome of an RNAi experiment and to avoid these unwanted off-target effects, an additional non-overlapping dsRNA can be used to down-regulate the same gene. However it has never been tested whether this approach is sufficient to reduce the risk of off-targets. Methodology We created a novel tool to analyse the occurance of off-target effects in Drosophila and we analyzed 99 randomly chosen genes. Principal Findings Here we show that nearly all genes contain non-overlapping internal sequences that do show overlap in a common off-target gene. Conclusion Based on our in silico findings, off-target effects should not be ignored and our presented on-line tool enables the identification of two RNA interference constructs, free of overlapping off-targets, from any gene of interest.
Collapse
Affiliation(s)
- Erwin Seinen
- Section of Radiation and Stress Cell Biology, Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Johannes G. M. Burgerhof
- Epidemiology, Faculty of Medical Sciences, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Ritsert C. Jansen
- Groningen Bioinformatics Centre, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Haren, The Netherlands
| | - Ody C. M. Sibon
- Section of Radiation and Stress Cell Biology, Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
45
|
Esmaeili F, Bamdad T, Ghasemi S. Stable suppression of gene expression by short interfering RNAs targeted to promoter in a mouse embryonal carcinoma stem cell line. In Vitro Cell Dev Biol Anim 2010; 46:834-40. [PMID: 20872183 DOI: 10.1007/s11626-010-9347-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 08/19/2010] [Indexed: 11/26/2022]
Abstract
RNA interference (RNAi) can induce gene silencing via two pathways: post-transcriptional gene silencing (PTGS) and transcriptional gene silencing (TGS). The mediators of gene inactivation in both pathways are 21-bp small interfering RNAs (siRNAs) generated from longer double-stranded RNA (dsRNA). PTGS involves siRNA-mediated targeting and degradation of mRNA. However, siRNAs induce TGS via DNA methylation at the targeted promoter. Synthetic siRNAs can induce loss of gene activity comparable to long dsRNA. The limitation of this method is that the transfected synthetic siRNA works for only a few days. In this study, we tested the RNAi response to siRNA (PTGS pathway) by using a plasmid containing an enhanced green fluorescent protein (eGFP) gene as a target as well as a plasmid creates siRNA transcript, in a form of a hairpin, against eGFP gene. To investigate TGS pathway via RNAi, we also used a plasmid creates hairpin siRNA transcript against pgk-1 promoter. The data presented here indicated long-lasting inhibition in expression of eGFP and puromycin genes, both under the control of the murine Pgk-1 promoter. However, Southern blot analysis showed no methylation in pgk-1 promoter.
Collapse
Affiliation(s)
- Fariba Esmaeili
- Department of Biology, Faculty of Basic Sciences, Shahr-e-kord University, Shahr-e-kord, Iran.
| | | | | |
Collapse
|
46
|
Abstract
DM1 (myotonic dystrophy type I) is a common form of muscular dystrophy that affects mainly adults. It is a disease that belongs to the group of defective RNA export diseases, since a major part of the pathogenic mechanism of the disease is the retention of the mutant transcripts in the cell nucleus. The presence of an expanded CUG trinucleotide repeat in the 3'-UTR (3'-untranslated region) of the DMPK (myotonic dystrophy protein kinase) gene causes the attraction of RNA-binding proteins by the nuclear-located mutant transcripts. As a result of the occupation of the RNA-binding proteins, there is defective mis-splicing of several cellular transcripts. This is believed to be a major pathogenic mechanism of the disease and any attempt to repair the activities of the RNA-binding proteins or target the mutant transcripts should be beneficial for the patients. Certain approaches have been described in the literature and they demonstrate progress in various directions. The purpose of the present review is to summarize the successful attempts to tackle the pathogenesis caused by nuclear retention of mutant transcripts in myotonic dystrophy and to discuss the possible gains from such approaches.
Collapse
|
47
|
Rao DD, Maples PB, Senzer N, Kumar P, Wang Z, Pappen BO, Yu Y, Haddock C, Jay C, Phadke AP, Chen S, Kuhn J, Dylewski D, Scott S, Monsma D, Webb C, Tong A, Shanahan D, Nemunaitis J. Enhanced target gene knockdown by a bifunctional shRNA: a novel approach of RNA interference. Cancer Gene Ther 2010; 17:780-91. [PMID: 20596090 DOI: 10.1038/cgt.2010.35] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RNA interference (RNAi) is a natural cellular regulatory process that inhibits gene expression by transcriptional, post-transcriptional and translational mechanisms. Synthetic approaches that emulate this process (small interfering RNA (siRNA), short hairpin RNA (shRNA)) have been shown to be similarly effective in this regard. We developed a novel 'bifunctional' RNAi strategy, which further optimizes target gene knockdown outcome. A bifunctional construct (bi-sh-STMN1) was generated against Stathmin1, a critical tubulin modulator that is overexpressed in human cancers. The bifunctional construct is postulated to concurrently repress the translation of the target mRNA (cleavage-independent, mRNA sequestration and degradation) and degrade (through RNase H-like cleavage) post-transcriptional mRNA through cleavage-dependent activities. Bi-sh-STMN1 showed enhanced potency and durability in parallel comparisons with conventional shRNA and siRNAs targeting the same sequence. Enhanced STMN1 protein knockdown by bi-sh-STMN1 was accompanied by target site cleavage at the mRNA level showed by the rapid amplification of complementary DNA ends (RACE) assay. Bi-sh-STMN1 also showed knockdown kinetics at the mRNA level consistent with its multieffector silencing mechanisms. The bifunctional shRNA is a highly effective and advantageous approach mediating RNAi at concentrations significantly lower than conventional shRNA or siRNA. These results support further evaluations.
Collapse
Affiliation(s)
- D D Rao
- Gradalis, Inc., Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mulders SAM, van Engelen BGM, Wieringa B, Wansink DG. Molecular therapy in myotonic dystrophy: focus on RNA gain-of-function. Hum Mol Genet 2010; 19:R90-7. [DOI: 10.1093/hmg/ddq161] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Schoser B, Timchenko L. Myotonic dystrophies 1 and 2: complex diseases with complex mechanisms. Curr Genomics 2010; 11:77-90. [PMID: 20885816 PMCID: PMC2874224 DOI: 10.2174/138920210790886844] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2009] [Revised: 11/22/2009] [Accepted: 11/25/2009] [Indexed: 11/22/2022] Open
Abstract
Two multi-system disorders, Myotonic Dystrophies type 1 and type 2 (DM1 and DM2), are complex neuromuscular diseases caused by an accumulation of expanded, non-coding RNAs, containing repetitive CUG and CCUG elements. Similarities of these mutations suggest similar mechanisms for both diseases. The expanded CUGn and CCUGn RNAs mainly target two RNA binding proteins, MBNL1 and CUGBP1, elevating levels of CUGBP1 and reducing levels of MBNL1. These alterations change processing of RNAs that are regulated by these proteins. Whereas overall toxicity of CUGn/CCUGn RNAs on RNA homeostasis in DM cells has been proven, the mechanisms which make these RNAs toxic remain illusive. A current view is that the toxicity of RNA CUGn and CCUGn is associated exclusively with global mis-splicing in DM patients. However, a growing number of new findings show that the expansion of CUGn and CCUGn RNAs mis-regulates several additional pathways in nuclei and cytoplasm of cells from patients with DM1 and DM2. The purpose of this review is to discuss the similarities and differences in the clinical presentation and molecular genetics of both diseases. We will also discuss the complexity of the molecular abnormalities in DM1 and DM2 caused by CUG and CCUG repeats and will summarize the outcomes of the toxicity of CUG and CCUG repeats.
Collapse
Affiliation(s)
- Benedikt Schoser
- Department of Neurology, Friedrich-Baur Institute, Ludwig-Maximilians-University, Ziemssenstr. 1a, Munich, Germany
| | - Lubov Timchenko
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
50
|
Epigenetic changes and non-coding expanded repeats. Neurobiol Dis 2010; 39:21-7. [PMID: 20171282 DOI: 10.1016/j.nbd.2010.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 02/08/2010] [Accepted: 02/09/2010] [Indexed: 12/31/2022] Open
Abstract
Many neurogenetic disorders are caused by unstable expansions of tandem repeats. Some of the causal mutations are located in non-protein-coding regions of genes. When pathologically expanded, these repeats can trigger focal epigenetic changes that repress the expression of the mutant allele. When the mutant gene is not repressed, the transcripts containing the expanded repeat can give rise to a toxic gain-of-function by the mutant RNA. These two mechanisms, heterochromatin-mediated gene repression and RNA dominance, produce a wide range of neurodevelopmental and neurodegenerative abnormalities. Here we review the mechanisms of gene dysregulation induced by non-coding repeat expansions, and early indications that some of these disorders may prove to be responsive to therapeutic intervention.
Collapse
|