Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nucleic Acids Res ; 49(16): 9479-9495, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34358321

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by a limited expansion of CGG repeats in the FMR1 gene. Degeneration of neurons in FXTAS cell models can be triggered by accumulation of polyglycine protein (FMRpolyG), a by-product of translation initiated upstream to the repeats. Specific aims of our work included testing if naphthyridine-based molecules could (i) block FMRpolyG synthesis by binding to CGG repeats in RNA, (ii) reverse pathological alterations in affected cells and (iii) preserve the content of FMRP, translated from the same FMR1 mRNA. We demonstrate that cyclic mismatch binding ligand CMBL4c binds to RNA structure formed by CGG repeats and attenuates translation of FMRpolyG and formation of nuclear inclusions in cells transfected with vectors expressing RNA with expanded CGG repeats. Moreover, our results indicate that CMBL4c delivery can reduce FMRpolyG-mediated cytotoxicity and apoptosis. Importantly, its therapeutic potential is also observed once the inclusions are already formed. We also show that CMBL4c-driven FMRpolyG loss is accompanied by partial FMRP reduction. As complete loss of FMRP induces FXS in children, future experiments should aim at evaluation of CMBL4c therapeutic intervention in differentiated tissues, in which FMRpolyG translation inhibition might outweigh adverse effects related to FMRP depletion.


Subject(s)
Ataxia/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Naphthyridines/pharmacology , Tremor/genetics , Trinucleotide Repeat Expansion/drug effects , Apoptosis/drug effects , Ataxia/drug therapy , Ataxia/pathology , Cell Proliferation/drug effects , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Syndrome/drug therapy , Fragile X Syndrome/pathology , HeLa Cells , Humans , Ligands , Neurons/drug effects , Neurons/pathology , Peptides/genetics , Protein Biosynthesis/drug effects , Surface Plasmon Resonance , Tremor/drug therapy , Tremor/pathology , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects , Trinucleotide Repeats/genetics
2.
Nucleic Acids Res ; 48(5): 2531-2543, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31965181

ABSTRACT

Expansion of an unstable CTG repeat in the 3'UTR of the DMPK gene causes Myotonic Dystrophy type 1 (DM1). CUG-expanded DMPK transcripts (CUGexp) sequester Muscleblind-like (MBNL) alternative splicing regulators in ribonuclear inclusions (foci), leading to abnormalities in RNA processing and splicing. To alleviate the burden of CUGexp, we tested therapeutic approach utilizing antisense oligonucleotides (AONs)-mediated DMPK splice-switching and degradation of mutated pre-mRNA. Experimental design involved: (i) skipping of selected constitutive exons to induce frameshifting and decay of toxic mRNAs by an RNA surveillance mechanism, and (ii) exclusion of the alternative exon 15 (e15) carrying CUGexp from DMPK mRNA. While first strategy failed to stimulate DMPK mRNA decay, exclusion of e15 enhanced DMPK nuclear export but triggered accumulation of potentially harmful spliced out pre-mRNA fragment containing CUGexp. Neutralization of this fragment with antisense gapmers complementary to intronic sequences preceding e15 failed to diminish DM1-specific spliceopathy due to AONs' chemistry-related toxicity. However, intronic gapmers alone reduced the level of DMPK mRNA and mitigated DM1-related cellular phenotypes including spliceopathy and nuclear foci. Thus, a combination of the correct chemistry and experimental approach should be carefully considered to design a safe AON-based therapeutic strategy for DM1.


Subject(s)
Alternative Splicing/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , Myotonin-Protein Kinase/genetics , Oligonucleotides, Antisense/therapeutic use , RNA Precursors/genetics , RNA Stability/genetics , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Exons/genetics , Humans , Myotonin-Protein Kinase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trinucleotide Repeat Expansion/genetics
3.
RNA Biol ; 15(1): 1-8, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28949831

ABSTRACT

Muscleblind-like (MBNL) proteins bind to hundreds of pre- and mature mRNAs to regulate their alternative splicing, alternative polyadenylation, stability and subcellular localization. Once MBNLs are withheld from transcript regulation, cellular machineries generate products inapt for precise embryonal/adult developmental tasks and myotonic dystrophy, a devastating multi-systemic genetic disorder, develops. We have recently demonstrated that all three MBNL paralogs are capable of fine-tuning cellular content of one of the three MBNL paralogs, MBNL1, by binding to the first coding exon (e1) of its pre-mRNA. Intriguingly, this autoregulatory feedback loop grounded on alternative splicing of e1 appears to play a crucial role in delaying the onset of myotonic dystrophy. Here, we describe this process in the context of other autoregulatory and regulatory loops that maintain the content and diverse functions of MBNL proteins at optimal level in health and disease, thus supporting the overall cellular homeostasis.


Subject(s)
Alternative Splicing/genetics , Myotonic Dystrophy/genetics , RNA-Binding Proteins/genetics , Exons/genetics , Feedback, Physiological , Gene Expression Regulation, Developmental , Humans , Myotonic Dystrophy/pathology , Polyadenylation/genetics , Protein Binding , RNA Precursors/genetics , RNA Splicing Factors/genetics
4.
Nucleic Acids Res ; 45(4): 1760-1775, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27903900

ABSTRACT

Muscleblind-like proteins (MBNLs) are regulators of RNA metabolism. During tissue differentiation the level of MBNLs increases, while their functional insufficiency plays a crucial role in myotonic dystrophy (DM). Deep sequencing of RNA molecules cross-linked to immunoprecipitated protein particles (CLIP-seq) revealed that MBNL1 binds to MBNL1 exon 1 (e1) encoding both the major part of 5΄UTR and an amino-terminal region of MBNL1 protein. We tested several hypotheses regarding the possible autoregulatory function of MBNL1 binding to its own transcript. Our data indicate that MBNLs induce skipping of e1 from precursor MBNL1 mRNA and that e1 exclusion may impact transcript association with polysomes and translation. Furthermore, e1-deficient protein isoform lacking the first two zinc fingers is highly unstable and its EGFP fusion protein has severely compromised splicing activity. We also show that MBNL1 can be transcribed from three different promoters and that the transcription initiation site determines the mode of e1 regulation. Taken together, we demonstrate that MBNL proteins control steady-state levels of MBNL1 through an interaction with e1 in its precursor mRNA. Insights from our study open a new avenue in therapies against DM based on manipulation of the transcription initiation site and e1 splicing of MBNL1 mRNA.


Subject(s)
Alternative Splicing , Exons , Gene Expression Regulation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Line , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Humans , Mice , Nucleic Acid Conformation , Protein Binding , Protein Stability , RNA Isoforms , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism
5.
Hum Mol Genet ; 24(17): 4971-83, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26082468

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.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Myotonic Dystrophy/genetics , RNA Interference , Animals , Cell Line , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression , Gene Order , Gene Silencing , Genetic Vectors/administration & dosage , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myotonic Dystrophy/therapy , Phenotype , RNA Splicing , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic , Transduction, Genetic , Transgenes
6.
Nucleic Acids Res ; 43(6): 3318-31, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25753670

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant multisystemic disorder caused by expansion of CTG triplet repeats in 3'-untranslated region of DMPK gene. The pathomechanism of DM1 is driven by accumulation of toxic transcripts containing expanded CUG repeats (CUG(exp)) in nuclear foci which sequester several factors regulating RNA metabolism, such as Muscleblind-like proteins (MBNLs). In this work, we utilized very short chemically modified antisense oligonucleotides composed exclusively of locked nucleic acids (all-LNAs) complementary to CUG repeats, as potential therapeutic agents against DM1. Our in vitro data demonstrated that very short, 8- or 10-unit all-LNAs effectively bound the CUG repeat RNA and prevented the formation of CUG(exp)/MBNL complexes. In proliferating DM1 cells as well as in skeletal muscles of DM1 mouse model the all-LNAs induced the reduction of the number and size of CUG(exp) foci and corrected MBNL-sensitive alternative splicing defects with high efficacy and specificity. The all-LNAs had low impact on the cellular level of CUG(exp)-containing transcripts and did not affect the expression of other transcripts with short CUG repeats. Our data strongly indicate that short all-LNAs complementary to CUG repeats are a promising therapeutic tool against DM1.


Subject(s)
Alternative Splicing , Myotonic Dystrophy/therapy , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , 3' Untranslated Regions , Animals , Base Sequence , Cell Line , Disease Models, Animal , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Transgenic , Mutant Proteins/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/genetics , Oligonucleotides/genetics , Oligonucleotides, Antisense/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Trinucleotide Repeat Expansion
7.
Nucleic Acids Res ; 42(17): 10873-87, 2014.
Article in English | MEDLINE | ID: mdl-25183524

ABSTRACT

Muscleblind-like (MBNL) proteins are key regulators of precursor and mature mRNA metabolism in mammals. Based on published and novel data, we explore models of tissue-specific MBNL interaction with RNA. We portray MBNL domains critical for RNA binding and splicing regulation, and the structure of MBNL's normal and pathogenic RNA targets, particularly in the context of myotonic dystrophy (DM), in which expanded CUG or CCUG repeat transcripts sequester several nuclear proteins including MBNLs. We also review the properties of MBNL/RNA complex, including recent data obtained from UV cross-linking and immunoprecipitation (CLIP-Seq), and discuss how this interaction shapes normal MBNL-dependent alternative splicing regulation. Finally, we review how this acquired knowledge about the pathogenic RNA structure and nature of MBNL sequestration can be translated into the design of therapeutic strategies against DM.


Subject(s)
Alternative Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , Protein Binding , Protein Structure, Tertiary , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics
8.
Nat Commun ; 4: 2044, 2013.
Article in English | MEDLINE | ID: mdl-23806903

ABSTRACT

The ability to control pre-mRNA splicing with small molecules could facilitate the development of therapeutics or cell-based circuits that control gene function. Myotonic dystrophy type 1 is caused by the dysregulation of alternative pre-mRNA splicing due to sequestration of muscleblind-like 1 protein (MBNL1) by expanded, non-coding r(CUG) repeats (r(CUG)(exp)). Here we report two small molecules that induce or ameliorate alternative splicing dysregulation. A thiophene-containing small molecule (1) inhibits the interaction of MBNL1 with its natural pre-mRNA substrates. Compound (2), a substituted naphthyridine, binds r(CUG)(exp) and displaces MBNL1. Structural models show that 1 binds MBNL1 in the Zn-finger domain and that 2 interacts with UU loops in r(CUG)(exp). This study provides a structural framework for small molecules that target MBNL1 by mimicking r(CUG)(exp) and shows that targeting MBNL1 causes dysregulation of alternative splicing, suggesting that MBNL1 is thus not a suitable therapeutic target for the treatment of myotonic dystrophy type 1.


Subject(s)
Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , RNA Precursors/genetics , RNA Splicing/genetics , Small Molecule Libraries/therapeutic use , Animals , Base Sequence , Exons/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , High-Throughput Screening Assays , Humans , Mice , Molecular Docking Simulation , Molecular Sequence Data , Plasmids/metabolism , Protein Binding/genetics , Protein Biosynthesis/drug effects , RNA/metabolism , RNA Precursors/metabolism , RNA Splicing/drug effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor, Insulin/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...