1
|
Hicks SM, Frias JA, Mishra SK, Scotti M, Muscato DR, Valero MC, Adams LM, Cleary JD, Nakamori M, Wang E, Berglund JA. Alternative splicing dysregulation across tissue and therapeutic approaches in a mouse model of myotonic dystrophy type 1. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102338. [PMID: 39391766 PMCID: PMC11465180 DOI: 10.1016/j.omtn.2024.102338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024]
Abstract
Myotonic dystrophy type 1 (DM1), the leading cause of adult-onset muscular dystrophy, is caused by a CTG repeat expansion. Expression of the repeat causes widespread alternative splicing (AS) defects and downstream pathogenesis, including significant skeletal muscle impacts. The HSA LR mouse model plays a significant role in therapeutic development. This mouse model features a transgene composed of approximately 220 interrupted CTG repeats, which results in skeletal muscle pathology that mirrors DM1. To better understand this model and the growing number of therapeutic approaches developed with it, we performed a meta-analysis of publicly available RNA sequencing data for AS changes across three widely examined skeletal muscles: quadriceps, gastrocnemius, and tibialis anterior. Our analysis demonstrated that transgene expression correlated with the extent of splicing dysregulation across these muscles from gastrocnemius (highest), quadriceps (medium), to tibialis anterior (lowest). We identified 95 splicing events consistently dysregulated across all examined datasets. Comparison of splicing rescue across seven therapeutic approaches showed a range of rescue across the 95 splicing events from the three muscle groups. This analysis contributes to our understanding of the HSA LR model and the growing number of therapeutic approaches currently in preclinical development for DM1.
Collapse
Affiliation(s)
- Sawyer M. Hicks
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Jesus A. Frias
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Subodh K. Mishra
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Marina Scotti
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - Derek R. Muscato
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - M. Carmen Valero
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - Leanne M. Adams
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - John D. Cleary
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eric Wang
- Center for NeuroGenetics and Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - J. Andrew Berglund
- Department of Biological Sciences, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
- The RNA Institute, College of Arts and Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| |
Collapse
|
2
|
Louis JM, Frias JA, Schroader JH, Jones LA, Davey EE, Lennon CD, Chacko J, Cleary JD, Berglund JA, Reddy K. Expression levels of core spliceosomal proteins modulate the MBNL-mediated spliceopathy in DM1. Hum Mol Genet 2024; 33:1873-1886. [PMID: 39180495 PMCID: PMC11540926 DOI: 10.1093/hmg/ddae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/26/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a heterogeneous multisystemic disease caused by a CTG repeat expansion in DMPK. Transcription of the expanded allele produces toxic CUG repeat RNA that sequesters the MBNL family of alternative splicing (AS) regulators into ribonuclear foci, leading to pathogenic mis-splicing. To identify genetic modifiers of toxic CUG RNA levels and the spliceopathy, we performed a genome-scale siRNA screen using an established HeLa DM1 repeat-selective screening platform. We unexpectedly identified core spliceosomal proteins as a new class of modifiers that rescue the spliceopathy in DM1. Modest knockdown of one of our top hits, SNRPD2, in DM1 fibroblasts and myoblasts, significantly reduces DMPK expression and partially rescues MBNL-regulated AS dysfunction. While the focus on the DM1 spliceopathy has centered around the MBNL proteins, our work reveals an unappreciated role for MBNL:spliceosomal protein stoichiometry in modulating the spliceopathy, revealing new biological and therapeutic avenues for DM1.
Collapse
Affiliation(s)
- Jiss M Louis
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Jesus A Frias
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Jacob H Schroader
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Lindsey A Jones
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Emily E Davey
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Claudia D Lennon
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Jacob Chacko
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - John D Cleary
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - J Andrew Berglund
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Kaalak Reddy
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| |
Collapse
|
3
|
Johnson SJ, Johnson HL, Powell RT, Stephan C, Stossi F, Cooper TA. Small Molecule Screening Identifies HSP90 as a Modifier of RNA Foci in Myotonic Dystrophy Type 1. Mol Cell Biol 2024:1-13. [PMID: 39415708 DOI: 10.1080/10985549.2024.2408025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disorder caused by a CTG triplet repeat expansion within the 3' untranslated region of the DMPK gene. Expression of the expanded allele generates RNA containing long tracts of CUG repeats (CUGexp RNA) that form hairpin structures and accumulate in nuclear RNA foci; however, the factors that control DMPK expression and the formation of CUGexp RNA foci remain largely unknown. We performed an unbiased small molecule screen in an immortalized human DM1 skeletal muscle myoblast cell line and identified HSP90 as a modifier of endogenous RNA foci. Small molecule inhibition of HSP90 leads to enhancement of RNA foci and upregulation of DMPK mRNA levels. Knockdown and overexpression of HSP90 in undifferentiated DM1 myoblasts validated the impact of HSP90 with upregulation and downregulation of DMPK mRNA, respectively. Furthermore, we identified p-STAT3 as a downstream mediator of HSP90 impacting levels of DMPK mRNA and RNA foci. Interestingly, differentiated cells exhibited an opposite effect of HSP90 inhibition displaying downregulation of DMPK mRNA through a mechanism independent of p-STAT3 involvement. This study has revealed a novel mediator for DMPK mRNA and foci regulation in DM1 cells with the potential to identify targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Sara J Johnson
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Hannah L Johnson
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Reid T Powell
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Clifford Stephan
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Fabio Stossi
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas A Cooper
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
4
|
Shorrock HK, Aliyeva A, Frias JA, DeMeo VA, Lennon CD, DeMeo CC, Mascorro AK, Shaughnessy S, Mazdiyasni H, Cleary JD, Reddy K, Vangaveti S, Shin DS, Berglund JA. CAG repeat-selective compounds reduce abundance of expanded CAG RNAs in patient cell and murine models of SCAs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.17.608349. [PMID: 39211226 PMCID: PMC11360937 DOI: 10.1101/2024.08.17.608349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Spinocerebellar ataxias (SCAs) are a genetically heterogenous group of devastating neurodegenerative conditions for which clinical care currently focuses on managing symptoms. Across these diseases there is an unmet need for therapies that address underlying disease mechanisms. We utilised the shared CAG repeat expansion mutation causative for a large subgroup of SCAs, to develop a novel disease-gene independent and mechanism agnostic small molecule screening approach to identify compounds with therapeutic potential across multiple SCAs. Using this approach, we identified the FDA approved microtubule inhibitor Colchicine and a novel CAG-repeat binding compound that reduce expression of disease associated transcripts across SCA1, 3 and 7 patient derived fibroblast lines and the Atxn1 154Q/2Q SCA1 mouse model in a repeat selective manner. Furthermore, our lead candidate rescues dysregulated alternative splicing in Atxn1 154Q/2Q mice. This work provides the first example of small molecules capable of targeting the underlying mechanism of disease across multiple CAG SCAs.
Collapse
|
5
|
Nagasawa R, Onizuka K, Komatsu KR, Miyashita E, Murase H, Ojima K, Ishikawa S, Ozawa M, Saito H, Nagatsugi F. Large-scale analysis of small molecule-RNA interactions using multiplexed RNA structure libraries. Commun Chem 2024; 7:98. [PMID: 38693284 DOI: 10.1038/s42004-024-01181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/17/2024] [Indexed: 05/03/2024] Open
Abstract
The large-scale analysis of small-molecule binding to diverse RNA structures is key to understanding the required interaction properties and selectivity for developing RNA-binding molecules toward RNA-targeted therapies. Here, we report a new system for performing the large-scale analysis of small molecule-RNA interactions using a multiplexed pull-down assay with RNA structure libraries. The system profiled the RNA-binding landscapes of G-clamp and thiazole orange derivatives, which recognizes an unpaired guanine base and are good probes for fluorescent indicator displacement (FID) assays, respectively. We discuss the binding preferences of these molecules based on their large-scale affinity profiles. In addition, we selected combinations of fluorescent indicators and different ranks of RNA based on the information and screened for RNA-binding molecules using FID. RNAs with high- and intermediate-rank RNA provided reliable results. Our system provides fundamental information about small molecule-RNA interactions and facilitates the discovery of novel RNA-binding molecules.
Collapse
Affiliation(s)
- Ryosuke Nagasawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Miyagi, 980-8578, Japan
| | - Kazumitsu Onizuka
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan.
- Department of Chemistry, Graduate School of Science, Tohoku University, Miyagi, 980-8578, Japan.
- Division for the Establishment of Frontier Sciences of Organization for Advanced Studies, Tohoku University, Miyagi, 980-8577, Japan.
| | - Kaoru R Komatsu
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Emi Miyashita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Hirotaka Murase
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan
| | - Kanna Ojima
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Miyagi, 980-8578, Japan
| | - Shunya Ishikawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Miyagi, 980-8578, Japan
| | - Mamiko Ozawa
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan
| | - Hirohide Saito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.
| | - Fumi Nagatsugi
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Miyagi, 980-8577, Japan.
- Department of Chemistry, Graduate School of Science, Tohoku University, Miyagi, 980-8578, Japan.
| |
Collapse
|
6
|
Shorrock HK, Lennon CD, Aliyeva A, Davey EE, DeMeo CC, Pritchard CE, Planco L, Velez JM, Mascorro-Huamancaja A, Shin DS, Cleary JD, Berglund JA. Widespread alternative splicing dysregulation occurs presymptomatically in CAG expansion spinocerebellar ataxias. Brain 2024; 147:486-504. [PMID: 37776516 PMCID: PMC10834251 DOI: 10.1093/brain/awad329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/31/2023] [Accepted: 09/03/2023] [Indexed: 10/02/2023] Open
Abstract
The spinocerebellar ataxias (SCAs) are a group of dominantly inherited neurodegenerative diseases, several of which are caused by CAG expansion mutations (SCAs 1, 2, 3, 6, 7 and 12) and more broadly belong to the large family of over 40 microsatellite expansion diseases. While dysregulation of alternative splicing is a well defined driver of disease pathogenesis across several microsatellite diseases, the contribution of alternative splicing in CAG expansion SCAs is poorly understood. Furthermore, despite extensive studies on differential gene expression, there remains a gap in our understanding of presymptomatic transcriptomic drivers of disease. We sought to address these knowledge gaps through a comprehensive study of 29 publicly available RNA-sequencing datasets. We identified that dysregulation of alternative splicing is widespread across CAG expansion mouse models of SCAs 1, 3 and 7. These changes were detected presymptomatically, persisted throughout disease progression, were repeat length-dependent, and were present in brain regions implicated in SCA pathogenesis including the cerebellum, pons and medulla. Across disease progression, changes in alternative splicing occurred in genes that function in pathways and processes known to be impaired in SCAs, such as ion channels, synaptic signalling, transcriptional regulation and the cytoskeleton. We validated several key alternative splicing events with known functional consequences, including Trpc3 exon 9 and Kcnma1 exon 23b, in the Atxn1154Q/2Q mouse model. Finally, we demonstrated that alternative splicing dysregulation is responsive to therapeutic intervention in CAG expansion SCAs with Atxn1 targeting antisense oligonucleotide rescuing key splicing events. Taken together, these data demonstrate that widespread presymptomatic dysregulation of alternative splicing in CAG expansion SCAs may contribute to disease onset, early neuronal dysfunction and may represent novel biomarkers across this devastating group of neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Claudia D Lennon
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
| | - Asmer Aliyeva
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
- Department of Biology, University at Albany—SUNY, Albany, NY 12222, USA
| | - Emily E Davey
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
| | - Cristina C DeMeo
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
| | | | - Lori Planco
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
| | - Jose M Velez
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
- Department of Biology, University at Albany—SUNY, Albany, NY 12222, USA
| | | | - Damian S Shin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208, USA
| | - John D Cleary
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
| | - J Andrew Berglund
- The RNA Institute, University at Albany—SUNY, Albany, NY 12222, USA
- Department of Biology, University at Albany—SUNY, Albany, NY 12222, USA
| |
Collapse
|
7
|
Nakamori M. Expanded‐repeat‐RNA‐mediated disease mechanisms in myotonic dystrophy. NEUROLOGY AND CLINICAL NEUROSCIENCE 2024; 12:16-23. [DOI: 10.1111/ncn3.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/05/2022] [Indexed: 01/04/2025]
Abstract
AbstractMyotonic dystrophy (DM) is the most common muscular dystrophy in adults, affecting skeletal muscle as well as cardiac and smooth muscle. Furthermore, involvement of the central nervous system, endocrine organs, and eyes is often seen, with debilitating consequences. The condition is an autosomal‐dominant inherited genetic disease caused by abnormal genomic expansion of tandem repeats. Myotonic dystrophy type 1 (DM1) results from expansion of a CTG repeat in the 3′‐untranslated region of the gene encoding dystrophia myotonica‐protein kinase (DMPK), whereas myotonic dystrophy type 2 (DM2) is caused by expansion of a CCTG repeat in the first intron of the gene encoding CCHC‐type zinc finger nucleic acid‐binding protein (CNBP). Both types of DM exhibit abnormal mRNA transcribed from the mutated gene containing expanded repeats, which exert toxic gain‐of‐function effects on various proteins involved in cellular processes such as alternative splicing, signaling pathways, and cellular senescence. The present review discusses the expanded‐repeat‐RNA‐mediated molecular pathomechanisms in DM.
Collapse
Affiliation(s)
- Masayuki Nakamori
- Department of Neurology Osaka University Graduate School of Medicine Osaka Japan
| |
Collapse
|
8
|
Puri D, Sharma S, Samaddar S, Ravivarma S, Banerjee S, Ghosh-Roy A. Muscleblind-1 interacts with tubulin mRNAs to regulate the microtubule cytoskeleton in C. elegans mechanosensory neurons. PLoS Genet 2023; 19:e1010885. [PMID: 37603562 PMCID: PMC10470942 DOI: 10.1371/journal.pgen.1010885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 08/31/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023] Open
Abstract
Regulation of the microtubule cytoskeleton is crucial for the development and maintenance of neuronal architecture, and recent studies have highlighted the significance of regulated RNA processing in the establishment and maintenance of neural circuits. In a genetic screen conducted using mechanosensory neurons of C. elegans, we identified a mutation in muscleblind-1/mbl-1 as a suppressor of loss of kinesin-13 family microtubule destabilizing factor klp-7. Muscleblind-1(MBL-1) is an RNA-binding protein that regulates the splicing, localization, and stability of RNA. Our findings demonstrate that mbl-1 is required cell-autonomously for axon growth and proper synapse positioning in the posterior lateral microtubule (PLM) neuron. Loss of mbl-1 leads to increased microtubule dynamics and mixed orientation of microtubules in the anterior neurite of PLM. These defects are also accompanied by abnormal axonal transport of the synaptic protein RAB-3 and reduction of gentle touch sensation in mbl-1 mutant. Our data also revealed that mbl-1 is genetically epistatic to mec-7 (β tubulin) and mec-12 (α tubulin) in regulating axon growth. Furthermore, mbl-1 is epistatic to sad-1, an ortholog of BRSK/Brain specific-serine/threonine kinase and a known regulator of synaptic machinery, for synapse formation at the correct location of the PLM neurite. Notably, the immunoprecipitation of MBL-1 resulted in the co-purification of mec-7, mec-12, and sad-1 mRNAs, suggesting a direct interaction between MBL-1 and these transcripts. Additionally, mbl-1 mutants exhibited reduced levels and stability of mec-7 and mec-12 transcripts. Our study establishes a previously unknown link between RNA-binding proteins and cytoskeletal machinery, highlighting their crucial roles in the development and maintenance of the nervous system.
Collapse
Affiliation(s)
- Dharmendra Puri
- National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Sunanda Sharma
- National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Sarbani Samaddar
- National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Sruthy Ravivarma
- National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | - Sourav Banerjee
- National Brain Research Centre, Manesar, Gurgaon, Haryana, India
| | | |
Collapse
|
9
|
Gibaut QR, Bush JA, Tong Y, Baisden JT, Taghavi A, Olafson H, Yao X, Childs-Disney JL, Wang ET, Disney MD. Transcriptome-Wide Studies of RNA-Targeted Small Molecules Provide a Simple and Selective r(CUG) exp Degrader in Myotonic Dystrophy. ACS CENTRAL SCIENCE 2023; 9:1342-1353. [PMID: 37521782 PMCID: PMC10375898 DOI: 10.1021/acscentsci.2c01223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Indexed: 08/01/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is caused by a highly structured RNA repeat expansion, r(CUG)exp, harbored in the 3' untranslated region (3' UTR) of dystrophia myotonica protein kinase (DMPK) mRNA and drives disease through a gain-of-function mechanism. A panel of low-molecular-weight fragments capable of reacting with RNA upon UV irradiation was studied for cross-linking to r(CUG)expin vitro, affording perimidin-2-amine diazirine (1) that bound to r(CUG)exp. The interactions between the small molecule and RNA were further studied by nuclear magnetic resonance (NMR) spectroscopy and molecular modeling. Binding of 1 in DM1 myotubes was profiled transcriptome-wide, identifying 12 transcripts including DMPK that were bound by 1. Augmenting the functionality of 1 with cleaving capability created a chimeric degrader that specifically targets r(CUG)exp for elimination. The degrader broadly improved DM1-associated defects as assessed by RNA-seq, while having limited effects on healthy myotubes. This study (i) provides a platform to investigate molecular recognition of ligands directly in disease-affected cells; (ii) illustrates that RNA degraders can be more specific than the binders from which they are derived; and (iii) suggests that repeating transcripts can be selectively degraded due to the presence of multiple ligand binding sites.
Collapse
Affiliation(s)
- Quentin
M. R. Gibaut
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jessica A. Bush
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Yuquan Tong
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jared T. Baisden
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Amirhossein Taghavi
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Hailey Olafson
- Center
for NeuroGenetics, University of Florida, Gainesville, Florida 32610, United States
- Department
of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Xiyuan Yao
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jessica L. Childs-Disney
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Eric T. Wang
- Center
for NeuroGenetics, University of Florida, Gainesville, Florida 32610, United States
- Department
of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Matthew D. Disney
- The
Department of Chemistry, UF Scripps Biomedical
Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| |
Collapse
|
10
|
Almeida CF, Robriquet F, Vetter TA, Huang N, Neinast R, Hernandez-Rosario L, Rajakumar D, Arnold WD, McBride KL, Flanigan KM, Weiss RB, Wein N. Promising AAV.U7snRNAs vectors targeting DMPK improve DM1 hallmarks in patient-derived cell lines. Front Cell Dev Biol 2023; 11:1181040. [PMID: 37397246 PMCID: PMC10309041 DOI: 10.3389/fcell.2023.1181040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/18/2023] [Indexed: 07/04/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults and affects mainly the skeletal muscle, heart, and brain. DM1 is caused by a CTG repeat expansion in the 3'UTR region of the DMPK gene that sequesters muscleblind-like proteins, blocking their splicing activity and forming nuclear RNA foci. Consequently, many genes have their splicing reversed to a fetal pattern. There is no treatment for DM1, but several approaches have been explored, including antisense oligonucleotides (ASOs) aiming to knock down DMPK expression or bind to the CTGs expansion. ASOs were shown to reduce RNA foci and restore the splicing pattern. However, ASOs have several limitations and although being safe treated DM1 patients did not demonstrate improvement in a human clinical trial. AAV-based gene therapies have the potential to overcome such limitations, providing longer and more stable expression of antisense sequences. In the present study, we designed different antisense sequences targeting exons 5 or 8 of DMPK and the CTG repeat tract aiming to knock down DMPK expression or promote steric hindrance, respectively. The antisense sequences were inserted in U7snRNAs, which were then vectorized in AAV8 particles. Patient-derived myoblasts treated with AAV8. U7snRNAs showed a significant reduction in the number of RNA foci and re-localization of muscle-blind protein. RNA-seq analysis revealed a global splicing correction in different patient-cell lines, without alteration in DMPK expression.
Collapse
Affiliation(s)
- Camila F. Almeida
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Florence Robriquet
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Tatyana A. Vetter
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Nianyuan Huang
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Reid Neinast
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - Dhanarajan Rajakumar
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
| | - W. David Arnold
- Department of Neurology, The Ohio State University, Columbus, OH, United States
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO, United States
| | - Kim L. McBride
- Center for Cardiovascular Research, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Kevin M. Flanigan
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Neurology, The Ohio State University, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Robert B. Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Nicolas Wein
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Mishra SK, Hicks SM, Frias JA, Vangaveti S, Nakamori M, Cleary JD, Reddy K, Berglund JA. Quercetin selectively reduces expanded repeat RNA levels in models of myotonic dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526846. [PMID: 36778282 PMCID: PMC9915578 DOI: 10.1101/2023.02.02.526846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Myotonic dystrophy is a multisystemic neuromuscular disease caused by either a CTG repeat expansion in DMPK (DM1) or a CCTG repeat expansion in CNBP (DM2). Transcription of the expanded alleles produces toxic gain-of-function RNA that sequester the MBNL family of alternative splicing regulators into ribonuclear foci, leading to pathogenic mis-splicing. There are currently no approved treatments that target the root cause of disease which is the production of the toxic expansion RNA molecules. In this study, using our previously established HeLa DM1 repeat selective screening platform, we identified the natural product quercetin as a selective modulator of toxic RNA levels. Quercetin treatment selectively reduced toxic RNA levels and rescued MBNL dependent mis-splicing in DM1 and DM2 patient derived cell lines and in the HSALR transgenic DM1 mouse model where rescue of myotonia was also observed. Based on our data and its safety profile for use in humans, we have identified quercetin as a priority disease-targeting therapeutic lead for clinical evaluation for the treatment of DM1 and DM2.
Collapse
Affiliation(s)
- Subodh K. Mishra
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Sawyer M. Hicks
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jesus A. Frias
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Sweta Vangaveti
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Masayuki Nakamori
- Department of Neurology, Osaka University Graduate School of Medicine; Osaka, Japan, 565-0871
| | - John D. Cleary
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Kaalak Reddy
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - J. Andrew Berglund
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
12
|
Kawada R, Jonouchi T, Kagita A, Sato M, Hotta A, Sakurai H. Establishment of quantitative and consistent in vitro skeletal muscle pathological models of myotonic dystrophy type 1 using patient-derived iPSCs. Sci Rep 2023; 13:94. [PMID: 36631509 PMCID: PMC9834395 DOI: 10.1038/s41598-022-26614-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is caused by expanded CTG repeats (CTGexp) in the dystrophia myotonica protein kinase (DMPK) gene, and the transcription products, expanded CUG repeats, sequester muscleblind like splicing regulator 1 (MBNL1), resulting in the nuclear MBNL1 aggregation in the DM1 cells. Loss of MBNL1 function is the pivotal mechanism underlying the pathogenesis of DM1. To develop therapeutics for DM1, proper human in vitro models based on the pathologic mechanism of DM1 are required. In this study, we established robust in vitro skeletal muscle cell models of DM1 with patient-derived induced pluripotent stem cells (iPSCs) using the MyoD1-induced system and iPSCs-derived muscle stem cell (iMuSC) differentiation system. Our newly established DM1 models enable simple quantitative evaluation of nuclear MBNL1 aggregation and the downstream splicing defects. Quantitative analyses using the MyoD1-induced myotubes showed that CTGexp-deleted DM1 skeletal myotubes exhibited a reversal of MBNL1-related pathologies, and antisense oligonucleotide treatment recovered these disease phenotypes in the DM1-iPSCs-derived myotubes. Furthermore, iMuSC-derived myotubes exhibited higher maturity than the MyoD1-induced myotubes, which enabled us to recapitulate the SERCA1 splicing defect in the DM1-iMuSC-derived myotubes. Our quantitative and reproducible in vitro models for DM1 established using human iPSCs are promising for drug discovery against DM1.
Collapse
Affiliation(s)
- Ryu Kawada
- grid.258799.80000 0004 0372 2033Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan ,grid.419836.10000 0001 2162 3360Discovery Research Laboratories, Taisho Pharmaceutical Co., Ltd., Saitama, 331-9530 Japan
| | - Tatsuya Jonouchi
- grid.258799.80000 0004 0372 2033Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
| | - Akihiro Kagita
- grid.258799.80000 0004 0372 2033Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
| | - Masae Sato
- grid.258799.80000 0004 0372 2033Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
| | - Akitsu Hotta
- grid.258799.80000 0004 0372 2033Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan.
| |
Collapse
|
13
|
Gibaut QMR, Akahori Y, Bush JA, Taghavi A, Tanaka T, Aikawa H, Ryan LS, Paegel BM, Disney MD. Study of an RNA-Focused DNA-Encoded Library Informs Design of a Degrader of a r(CUG) Repeat Expansion. J Am Chem Soc 2022; 144:21972-21979. [PMID: 36399603 PMCID: PMC9878440 DOI: 10.1021/jacs.2c08883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A solid-phase DNA-encoded library (DEL) was studied for binding the RNA repeat expansion r(CUG)exp, the causative agent of the most common form of adult-onset muscular dystrophy, myotonic dystrophy type 1 (DM1). A variety of uncharged and novel RNA binders were identified to selectively bind r(CUG)exp by using a two-color flow cytometry screen. The cellular activity of one binder was augmented by attaching it with a module that directly cleaves r(CUG)exp. In DM1 patient-derived muscle cells, the compound specifically bound r(CUG)exp and allele-specifically eliminated r(CUG)exp, improving disease-associated defects. The approaches herein can be used to identify and optimize ligands and bind RNA that can be further augmented for functionality including degradation.
Collapse
Affiliation(s)
- Quentin M. R. Gibaut
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Yoshihiro Akahori
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jessica A. Bush
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Amirhossein Taghavi
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Toru Tanaka
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Haruo Aikawa
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Lucas S. Ryan
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Brian M. Paegel
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States; Department of Chemistry and Pharmaceutical Sciences, University of California, Irvine, California 92617, United States
| | - Matthew D. Disney
- Department of Chemistry, UF Scripps Biomedical Research and The Scripps Research Institute, Jupiter, Florida 33458, United States
| |
Collapse
|
14
|
Development of Therapeutic Approaches for Myotonic Dystrophies Type 1 and Type 2. Int J Mol Sci 2022; 23:ijms231810491. [PMID: 36142405 PMCID: PMC9499601 DOI: 10.3390/ijms231810491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Myotonic Dystrophies type 1 (DM1) and type 2 (DM2) are complex multisystem diseases without disease-based therapies. These disorders are caused by the expansions of unstable CTG (DM1) and CCTG (DM2) repeats outside of the coding regions of the disease genes: DMPK in DM1 and CNBP in DM2. Multiple clinical and molecular studies provided a consensus for DM1 pathogenesis, showing that the molecular pathophysiology of DM1 is associated with the toxicity of RNA CUG repeats, which cause multiple disturbances in RNA metabolism in patients' cells. As a result, splicing, translation, RNA stability and transcription of multiple genes are misregulated in DM1 cells. While mutant CCUG repeats are the main cause of DM2, additional factors might play a role in DM2 pathogenesis. This review describes current progress in the translation of mechanistic knowledge in DM1 and DM2 to clinical trials, with a focus on the development of disease-specific therapies for patients with adult forms of DM1 and congenital DM1 (CDM1).
Collapse
|
15
|
Schroader JH, Jones LA, Meng R, Shorrock HK, Richardson J, Shaughnessy S, Lin Q, Begley T, Berglund J, Fuchs G, Handley M, Reddy K. Disease-associated inosine misincorporation into RNA hinders translation. Nucleic Acids Res 2022; 50:9306-9318. [PMID: 35979951 PMCID: PMC9458462 DOI: 10.1093/nar/gkac709] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/08/2022] [Accepted: 08/06/2022] [Indexed: 12/24/2022] Open
Abstract
Failure to prevent accumulation of the non-canonical nucleotide inosine triphosphate (ITP) by inosine triphosphate pyrophosphatase (ITPase) during nucleotide synthesis results in misincorporation of inosine into RNA and can cause severe and fatal developmental anomalies in humans. While the biochemical activity of ITPase is well understood, the pathogenic basis of ITPase deficiency and the molecular and cellular consequences of ITP misincorporation into RNA remain cryptic. Here, we demonstrate that excess ITP in the nucleotide pool during in vitro transcription results in T7 polymerase-mediated inosine misincorporation in luciferase RNA. In vitro translation of inosine-containing luciferase RNA reduces resulting luciferase activity, which is only partly explained by reduced abundance of the luciferase protein produced. Using Oxford Nanopore Direct RNA sequencing, we reveal inosine misincorporation to be stochastic but biased largely towards misincorporation in place of guanosine, with evidence for misincorporation also in place of cytidine, adenosine and uridine. Inosine misincorporation into RNA is also detected in Itpa-null mouse embryonic heart tissue as an increase in relative variants compared with the wild type using Illumina RNA sequencing. By generating CRISPR/Cas9 rat H9c2 Itpa-null cardiomyoblast cells, we validate a translation defect in cells that accumulate inosine within endogenous RNA. Furthermore, we observe hindered cellular translation of transfected luciferase RNA containing misincorporated inosine in both wild-type and Itpa-null cells. We therefore conclude that inosine misincorporation into RNA perturbs translation, thus providing mechanistic insight linking ITPase deficiency, inosine accumulation and pathogenesis.
Collapse
Affiliation(s)
| | | | - Ryan Meng
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Hannah K Shorrock
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jared I Richardson
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA,Department of Biochemistry and Molecular Biology, Center for NeuroGenetics, University of Florida, Gainesville, FL 32611, USA
| | - Sharon M Shaughnessy
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Qishan Lin
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA,RNA Epitranscriptomics & Proteomics Resource, University at Albany, Albany, NY 12222, USA
| | - Thomas J Begley
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA,Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA,RNA Epitranscriptomics & Proteomics Resource, University at Albany, Albany, NY 12222, USA
| | - J Andrew Berglund
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA,Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA,Department of Biochemistry and Molecular Biology, Center for NeuroGenetics, University of Florida, Gainesville, FL 32611, USA
| | - Gabriele Fuchs
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA,Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Mark T Handley
- Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Kaalak Reddy
- *To whom correspondence should be addressed. Tel: +1 518 442 1464;
| |
Collapse
|
16
|
Molecular Therapies for Myotonic Dystrophy Type 1: From Small Drugs to Gene Editing. Int J Mol Sci 2022; 23:ijms23094622. [PMID: 35563013 PMCID: PMC9101876 DOI: 10.3390/ijms23094622] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy affecting many different body tissues, predominantly skeletal and cardiac muscles and the central nervous system. The expansion of CTG repeats in the DM1 protein-kinase (DMPK) gene is the genetic cause of the disease. The pathogenetic mechanisms are mainly mediated by the production of a toxic expanded CUG transcript from the DMPK gene. With the availability of new knowledge, disease models, and technical tools, much progress has been made in the discovery of altered pathways and in the potential of therapeutic intervention, making the path to the clinic a closer reality. In this review, we describe and discuss the molecular therapeutic strategies for DM1, which are designed to directly target the CTG genomic tract, the expanded CUG transcript or downstream signaling molecules.
Collapse
|
17
|
Dastidar S, Majumdar D, Tipanee J, Singh K, Klein AF, Furling D, Chuah MK, VandenDriessche T. Comprehensive transcriptome-wide analysis of spliceopathy correction of myotonic dystrophy using CRISPR-Cas9 in iPSCs-derived cardiomyocytes. Mol Ther 2022; 30:75-91. [PMID: 34371182 PMCID: PMC8753376 DOI: 10.1016/j.ymthe.2021.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 07/01/2021] [Accepted: 07/26/2021] [Indexed: 01/07/2023] Open
Abstract
CTG repeat expansion (CTGexp) is associated with aberrant alternate splicing that contributes to cardiac dysfunction in myotonic dystrophy type 1 (DM1). Excision of this CTGexp repeat using CRISPR-Cas resulted in the disappearance of punctate ribonuclear foci in cardiomyocyte-like cells derived from DM1-induced pluripotent stem cells (iPSCs). This was associated with correction of the underlying spliceopathy as determined by RNA sequencing and alternate splicing analysis. Certain genes were of particular interest due to their role in cardiac development, maturation, and function (TPM4, CYP2J2, DMD, MBNL3, CACNA1H, ROCK2, ACTB) or their association with splicing (SMN2, GCFC2, MBNL3). Moreover, while comparing isogenic CRISPR-Cas9-corrected versus non-corrected DM1 cardiomyocytes, a prominent difference in the splicing pattern for a number of candidate genes was apparent pertaining to genes that are associated with cardiac function (TNNT, TNNT2, TTN, TPM1, SYNE1, CACNA1A, MTMR1, NEBL, TPM1), cellular signaling (NCOR2, CLIP1, LRRFIP2, CLASP1, CAMK2G), and other DM1-related genes (i.e., NUMA1, MBNL2, LDB3) in addition to the disease-causing DMPK gene itself. Subsequent validation using a selected gene subset, including MBNL1, MBNL2, INSR, ADD3, and CRTC2, further confirmed correction of the spliceopathy following CTGexp repeat excision. To our knowledge, the present study provides the first comprehensive unbiased transcriptome-wide analysis of the differential splicing landscape in DM1 patient-derived cardiac cells after excision of the CTGexp repeat using CRISPR-Cas9, showing reversal of the abnormal cardiac spliceopathy in DM1.
Collapse
Affiliation(s)
- Sumitava Dastidar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Debanjana Majumdar
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jaitip Tipanee
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Kshitiz Singh
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Arnaud F. Klein
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Denis Furling
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marinee K. Chuah
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Marinee K. Chuah, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium,Center for Molecular & Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, 3000 Leuven, Belgium,Corresponding author: Thierry VandenDriessche, Department of Gene Therapy & Regenerative Medicine, Vrije Universiteit Brussel, 1090 Brussels, Belgium.
| |
Collapse
|
18
|
Disrupting the Molecular Pathway in Myotonic Dystrophy. Int J Mol Sci 2021; 22:ijms222413225. [PMID: 34948025 PMCID: PMC8708683 DOI: 10.3390/ijms222413225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/26/2023] Open
Abstract
Myotonic dystrophy is the most common muscular dystrophy in adults. It consists of two forms: type 1 (DM1) and type 2 (DM2). DM1 is associated with a trinucleotide repeat expansion mutation, which is transcribed but not translated into protein. The mutant RNA remains in the nucleus, which leads to a series of downstream abnormalities. DM1 is widely considered to be an RNA-based disorder. Thus, we consider three areas of the RNA pathway that may offer targeting opportunities to disrupt the production, stability, and degradation of the mutant RNA.
Collapse
|
19
|
Liu J, Guo ZN, Yan XL, Yang Y, Huang S. Brain Pathogenesis and Potential Therapeutic Strategies in Myotonic Dystrophy Type 1. Front Aging Neurosci 2021; 13:755392. [PMID: 34867280 PMCID: PMC8634727 DOI: 10.3389/fnagi.2021.755392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy that affects multiple systems including the muscle and heart. The mutant CTG expansion at the 3'-UTR of the DMPK gene causes the expression of toxic RNA that aggregate as nuclear foci. The foci then interfere with RNA-binding proteins, affecting hundreds of mis-spliced effector genes, leading to aberrant alternative splicing and loss of effector gene product functions, ultimately resulting in systemic disorders. In recent years, increasing clinical, imaging, and pathological evidence have indicated that DM1, though to a lesser extent, could also be recognized as true brain diseases, with more and more researchers dedicating to develop novel therapeutic tools dealing with it. In this review, we summarize the current advances in the pathogenesis and pathology of central nervous system (CNS) deficits in DM1, intervention measures currently being investigated are also highlighted, aiming to promote novel and cutting-edge therapeutic investigations.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Zhen-Ni Guo
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Xiu-Li Yan
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| | - Shuo Huang
- Department of Neurology, Stroke Center & Clinical Trial and Research Center for Stroke, The First Hospital of Jilin University, Changchun, China
- China National Comprehensive Stroke Center, Changchun, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China
| |
Collapse
|
20
|
Hinman MN, Richardson JI, Sockol RA, Aronson ED, Stednitz SJ, Murray KN, Berglund JA, Guillemin K. Zebrafish mbnl mutants model physical and molecular phenotypes of myotonic dystrophy. Dis Model Mech 2021; 14:dmm045773. [PMID: 34125183 PMCID: PMC8246264 DOI: 10.1242/dmm.045773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
The muscleblind RNA-binding proteins (MBNL1, MBNL2 and MBNL3) are highly conserved across vertebrates and are important regulators of RNA alternative splicing. Loss of MBNL protein function through sequestration by CUG or CCUG RNA repeats is largely responsible for the phenotypes of the human genetic disorder myotonic dystrophy (DM). We generated the first stable zebrafish (Danio rerio) models of DM-associated MBNL loss of function through mutation of the three zebrafish mbnl genes. In contrast to mouse models, zebrafish double and triple homozygous mbnl mutants were viable to adulthood. Zebrafish mbnl mutants displayed disease-relevant physical phenotypes including decreased body size and impaired movement. They also exhibited widespread alternative splicing changes, including the misregulation of many DM-relevant exons. Physical and molecular phenotypes were more severe in compound mbnl mutants than in single mbnl mutants, suggesting partially redundant functions of Mbnl proteins. The high fecundity and larval optical transparency of this complete series of zebrafish mbnl mutants will make them useful for studying DM-related phenotypes and how individual Mbnl proteins contribute to them, and for testing potential therapeutics. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Melissa N. Hinman
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Jared I. Richardson
- RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
- Department of Biochemistry and Molecular Biology, Center for NeuroGenetics, University of Florida, Gainesville, FL 32611, USA
| | - Rose A. Sockol
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Eliza D. Aronson
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Sarah J. Stednitz
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - Katrina N. Murray
- Zebrafish International Resource Center, University of Oregon, Eugene, OR 97403, USA
| | - J. Andrew Berglund
- RNA Institute, State University of New York at Albany, Albany, NY 12222, USA
- Department of Biochemistry and Molecular Biology, Center for NeuroGenetics, University of Florida, Gainesville, FL 32611, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
- Humans and the Microbiome Program, CIFAR, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
21
|
Schwartz JL, Jones KL, Yeo GW. Repeat RNA expansion disorders of the nervous system: post-transcriptional mechanisms and therapeutic strategies. Crit Rev Biochem Mol Biol 2020; 56:31-53. [PMID: 33172304 PMCID: PMC8192115 DOI: 10.1080/10409238.2020.1841726] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dozens of incurable neurological disorders result from expansion of short repeat sequences in both coding and non-coding regions of the transcriptome. Short repeat expansions underlie microsatellite repeat expansion (MRE) disorders including myotonic dystrophy (DM1, CUG50–3,500 in DMPK; DM2, CCTG75–11,000 in ZNF9), fragile X tremor ataxia syndrome (FXTAS, CGG50–200 in FMR1), spinal bulbar muscular atrophy (SBMA, CAG40–55 in AR), Huntington’s disease (HD, CAG36–121 in HTT), C9ORF72-amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD and C9-ALS/FTD, GGGGCC in C9ORF72), and many others, like ataxias. Recent research has highlighted several mechanisms that may contribute to pathology in this heterogeneous class of neurological MRE disorders – bidirectional transcription, intranuclear RNA foci, and repeat associated non-AUG (RAN) translation – which are the subject of this review. Additionally, many MRE disorders share similar underlying molecular pathologies that have been recently targeted in experimental and preclinical contexts. We discuss the therapeutic potential of versatile therapeutic strategies that may selectively target disrupted RNA-based processes and may be readily adaptable for the treatment of multiple MRE disorders. Collectively, the strategies under consideration for treatment of multiple MRE disorders include reducing levels of toxic RNA, preventing RNA foci formation, and eliminating the downstream cellular toxicity associated with peptide repeats produced by RAN translation. While treatments are still lacking for the majority of MRE disorders, several promising therapeutic strategies have emerged and will be evaluated within this review.
Collapse
Affiliation(s)
- Joshua L Schwartz
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Krysten Leigh Jones
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Taylor K, Sobczak K. Intrinsic Regulatory Role of RNA Structural Arrangement in Alternative Splicing Control. Int J Mol Sci 2020; 21:ijms21145161. [PMID: 32708277 PMCID: PMC7404189 DOI: 10.3390/ijms21145161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Alternative splicing is a highly sophisticated process, playing a significant role in posttranscriptional gene expression and underlying the diversity and complexity of organisms. Its regulation is multilayered, including an intrinsic role of RNA structural arrangement which undergoes time- and tissue-specific alterations. In this review, we describe the principles of RNA structural arrangement and briefly decipher its cis- and trans-acting cellular modulators which serve as crucial determinants of biological functionality of the RNA structure. Subsequently, we engage in a discussion about the RNA structure-mediated mechanisms of alternative splicing regulation. On one hand, the impairment of formation of optimal RNA structures may have critical consequences for the splicing outcome and further contribute to understanding the pathomechanism of severe disorders. On the other hand, the structural aspects of RNA became significant features taken into consideration in the endeavor of finding potential therapeutic treatments. Both aspects have been addressed by us emphasizing the importance of ongoing studies in both fields.
Collapse
|
23
|
Khristich AN, Mirkin SM. On the wrong DNA track: Molecular mechanisms of repeat-mediated genome instability. J Biol Chem 2020; 295:4134-4170. [PMID: 32060097 PMCID: PMC7105313 DOI: 10.1074/jbc.rev119.007678] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Expansions of simple tandem repeats are responsible for almost 50 human diseases, the majority of which are severe, degenerative, and not currently treatable or preventable. In this review, we first describe the molecular mechanisms of repeat-induced toxicity, which is the connecting link between repeat expansions and pathology. We then survey alternative DNA structures that are formed by expandable repeats and review the evidence that formation of these structures is at the core of repeat instability. Next, we describe the consequences of the presence of long structure-forming repeats at the molecular level: somatic and intergenerational instability, fragility, and repeat-induced mutagenesis. We discuss the reasons for gender bias in intergenerational repeat instability and the tissue specificity of somatic repeat instability. We also review the known pathways in which DNA replication, transcription, DNA repair, and chromatin state interact and thereby promote repeat instability. We then discuss possible reasons for the persistence of disease-causing DNA repeats in the genome. We describe evidence suggesting that these repeats are a payoff for the advantages of having abundant simple-sequence repeats for eukaryotic genome function and evolvability. Finally, we discuss two unresolved fundamental questions: (i) why does repeat behavior differ between model systems and human pedigrees, and (ii) can we use current knowledge on repeat instability mechanisms to cure repeat expansion diseases?
Collapse
Affiliation(s)
| | - Sergei M Mirkin
- Department of Biology, Tufts University, Medford, Massachusetts 02155.
| |
Collapse
|