Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 34: 102024, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37744174

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a rare neuromuscular disease caused by a CTG repeat expansion in the DMPK gene that generates toxic RNA with a myriad of downstream alterations in RNA metabolism. A key consequence is the sequestration of alternative splicing regulatory proteins MBNL1/2 by expanded transcripts in the affected tissues. MBNL1/2 depletion interferes with a developmental alternative splicing switch that causes the expression of fetal isoforms in adults. Boosting the endogenous expression of MBNL proteins by inhibiting the natural translational repressors miR-23b and miR-218 has previously been shown to be a promising therapeutic approach. We designed antimiRs against both miRNAs with a phosphorodiamidate morpholino oligonucleotide (PMO) chemistry conjugated to cell-penetrating peptides (CPPs) to improve delivery to affected tissues. In DM1 cells, CPP-PMOs significantly increased MBNL1 levels. In some candidates, this was achieved using concentrations less than two orders of magnitude below the median toxic concentration, with up to 5.38-fold better therapeutic window than previous antagomiRs. In HSALR mice, intravenous injections of CPP-PMOs improve molecular, histopathological, and functional phenotypes, without signs of toxicity. Our findings place CPP-PMOs as promising antimiR candidates to overcome the treatment delivery challenge in DM1 therapy.

2.
Pharmaceutics ; 15(4)2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37111604

ABSTRACT

The symptoms of Myotonic Dystrophy Type 1 (DM1) are multi-systemic and life-threatening. The neuromuscular disorder is rooted in a non-coding CTG microsatellite expansion in the DM1 protein kinase (DMPK) gene that, upon transcription, physically sequesters the Muscleblind-like (MBNL) family of splicing regulator proteins. The high-affinity binding occurring between the proteins and the repetitions disallow MBNL proteins from performing their post-transcriptional splicing regulation leading to downstream molecular effects directly related to disease symptoms such as myotonia and muscle weakness. In this study, we build on previously demonstrated evidence showing that the silencing of miRNA-23b and miRNA-218 can increase MBNL1 protein in DM1 cells and mice. Here, we use blockmiR antisense technology in DM1 muscle cells, 3D mouse-derived muscle tissue, and in vivo mice to block the binding sites of these microRNAs in order to increase MBNL translation into protein without binding to microRNAs. The blockmiRs show therapeutic effects with the rescue of mis-splicing, MBNL subcellular localization, and highly specific transcriptomic expression. The blockmiRs are well tolerated in 3D mouse skeletal tissue inducing no immune response. In vivo, a candidate blockmiR also increases Mbnl1/2 protein and rescues grip strength, splicing, and histological phenotypes.

3.
Mol Ther Nucleic Acids ; 27: 1146-1155, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35282418

ABSTRACT

Myotonic dystrophy type 1 is a debilitating neuromuscular disease causing muscle weakness, myotonia, and cardiac dysfunction. The phenotypes are caused by muscleblind-like (MBNL) protein sequestration by toxic RNA in the DM1 protein kinase (DMPK) gene. DM1 patients exhibit a pathogenic number of repetitions in DMPK, which leads to downstream symptoms. Another disease characteristic is altered microRNA (miRNA) expression. It was previously shown that miR-23b regulates the translation of MBNL1 into protein. Antisense oligonucleotide (AON) treatment targeting this miRNA can improve disease symptoms. Here, we present a refinement of this strategy targeting a miR-23b binding site on the MBNL1 3' UTR in DM1 model cells and mice by using AONs called blockmiRs. BlockmiRs linked to novel cell-penetrating peptide chemistry showed an increase in MBNL1 protein in DM1 model cells and HSALR mice. They also showed an increase in muscle strength and significant rescue of downstream splicing and histological phenotypes in mice without disturbing the endogenous levels of other miR-23b target transcripts.

4.
Sci Rep ; 11(1): 19417, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593893

ABSTRACT

In Myotonic Dystrophy type 1 (DM1), a non-coding CTG repeats rare expansion disease; toxic double-stranded RNA hairpins sequester the RNA-binding proteins Muscleblind-like 1 and 2 (MBNL1 and 2) and trigger other DM1-related pathogenesis pathway defects. In this paper, we characterize four D-amino acid hexapeptides identified together with abp1, a peptide previously shown to stabilize CUG RNA in its single-stranded conformation. With the generalized sequence cpy(a/t)(q/w)e, these related peptides improved three MBNL-regulated exon inclusions in DM1-derived cells. Subsequent experiments showed that these compounds generally increased the relative expression of MBNL1 and its nuclear-cytoplasmic distribution, reduced hyperactivated autophagy, and increased the percentage of differentiated (Desmin-positive) cells in vitro. All peptides rescued atrophy of indirect flight muscles in a Drosophila model of the disease, and partially rescued muscle function according to climbing and flight tests. Investigation of their mechanism of action supports that all four compounds can bind to CUG repeats with slightly different association constant, but binding did not strongly influence the secondary structure of the toxic RNA in contrast to abp1. Finally, molecular modeling suggests a detailed view of the interactions of peptide-CUG RNA complexes useful in the chemical optimization of compounds.


Subject(s)
Myotonic Dystrophy/metabolism , Peptides/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Adolescent , Adult , Animals , Cells, Cultured , Drosophila , Female , Fibroblasts , Humans , Male , Protein Binding
5.
Mol Ther Nucleic Acids ; 26: 174-191, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34513303

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a rare neuromuscular disease caused by expansion of unstable CTG repeats in a non-coding region of the DMPK gene. CUG expansions in mutant DMPK transcripts sequester MBNL1 proteins in ribonuclear foci. Depletion of this protein is a primary contributor to disease symptoms such as muscle weakness and atrophy and myotonia, yet upregulation of endogenous MBNL1 levels may compensate for this sequestration. Having previously demonstrated that antisense oligonucleotides against miR-218 boost MBNL1 expression and rescue phenotypes in disease models, here we provide preclinical characterization of an antagomiR-218 molecule using the HSALR mouse model and patient-derived myotubes. In HSALR, antagomiR-218 reached 40-60 pM 2 weeks after injection, rescued molecular and functional phenotypes in a dose- and time-dependent manner, and showed a good toxicity profile after a single subcutaneous administration. In muscle tissue, antagomiR rescued the normal subcellular distribution of Mbnl1 and did not alter the proportion of myonuclei containing CUG foci. In patient-derived cells, antagomiR-218 improved defective fusion and differentiation and rescued up to 34% of the gene expression alterations found in the transcriptome of patient cells. Importantly, miR-218 was found to be upregulated in DM1 muscle biopsies, pinpointing this microRNA (miRNA) as a relevant therapeutic target.

6.
Sci Rep ; 11(1): 15287, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315987

ABSTRACT

Heart failure (HF) and the development of chronic kidney disease (CKD) have a direct association. Both can be cause and consequence of the other. Many factors are known, such as diabetes or hypertension, which can lead to the appearance and/or development of these two conditions. However, it is suspected that other factors, namely genetic ones, may explain the differences in the manifestation and progression of HF and CKD among patients. One candidate factor is Rph, a gene expressed in the nervous and excretory system in mammals and Drosophila, encoding a Rab small GTPase family effector protein implicated in vesicular trafficking. We found that Rph is expressed in the Drosophila heart, and the silencing of Rph gene expression in this organ had a strong impact in the organization of fibers and functional cardiac parameters. Specifically, we observed a significant increase in diastolic and systolic diameters of the heart tube, which is a phenotype that resembles dilated cardiomyopathy in humans. Importantly, we also show that silencing of Rabphilin (Rph) expression exclusively in the pericardial nephrocytes, which are part of the flies' excretory system, brings about a non-cell-autonomous effect on the Drosophila cardiac system. In summary, in this work, we demonstrate the importance of Rph in the fly cardiac system and how silencing Rph expression in nephrocytes affects the Drosophila cardiac system.


Subject(s)
Cardiomyopathy, Dilated/genetics , Drosophila Proteins/genetics , Gene Silencing , Nerve Tissue Proteins/genetics , Animals
7.
Mol Ther Nucleic Acids ; 21: 837-849, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32805487

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a chronically debilitating, rare genetic disease that originates from an expansion of a noncoding CTG repeat in the dystrophia myotonica protein kinase (DMPK) gene. The expansion becomes pathogenic when DMPK transcripts contain 50 or more repetitions due to the sequestration of the muscleblind-like (MBNL) family of proteins. Depletion of MBNLs causes alterations in splicing patterns in transcripts that contribute to clinical symptoms such as myotonia and muscle weakness and wasting. We previously found that microRNA (miR)-23b directly regulates MBNL1 in DM1 myoblasts and mice and that antisense technology ("antagomiRs") blocking this microRNA (miRNA) boosts MBNL1 protein levels. Here, we show the therapeutic effect over time in response to administration of antagomiR-23b as a treatment in human skeletal actin long repeat (HSALR) mice. Subcutaneous administration of antagomiR-23b upregulated the expression of MBNL1 protein and rescued splicing alterations, grip strength, and myotonia in a dose-dependent manner with long-lasting effects. Additionally, the effects of the treatment on grip strength and myotonia were still slightly notable after 45 days. The pharmacokinetic data obtained provide further evidence that miR-23b could be a valid therapeutic target for DM1.

8.
Dis Model Mech ; 13(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32680845

ABSTRACT

Drosophila nephrocytes share functional, structural and molecular similarities with human podocytes. It is known that podocytes express the rabphilin 3A (RPH3A)-RAB3A complex, and its expression is altered in mouse and human proteinuric disease. Furthermore, we previously identified a polymorphism that suggested a role for RPH3A protein in the development of urinary albumin excretion. As endocytosis and vesicle trafficking are fundamental pathways for nephrocytes, the objective of this study was to assess the role of the RPH3A orthologue in Drosophila, Rabphilin (Rph), in the structure and function of nephrocytes. We confirmed that Rph is required for the correct function of the endocytic pathway in pericardial Drosophila nephrocytes. Knockdown of Rph reduced the expression of the cubilin and stick and stones genes, which encode proteins that are involved in protein uptake and filtration. We also found that reduced Rph expression resulted in a disappearance of the labyrinthine channel structure and a reduction in the number of endosomes, which ultimately leads to changes in the number and volume of nephrocytes. Finally, we demonstrated that the administration of retinoic acid to IR-Rph nephrocytes rescued some altered aspects, such as filtration and molecular uptake, as well as the maintenance of cell fate. According to our data, Rph is crucial for nephrocyte filtration and reabsorption, and it is required for the maintenance of the ultrastructure, integrity and differentiation of the nephrocyte.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Podocytes/metabolism , Animals , Cell Lineage/drug effects , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/metabolism , Drosophila melanogaster/drug effects , Endocytosis/drug effects , Female , Humans , Larva/cytology , Larva/drug effects , Podocytes/cytology , Podocytes/drug effects , Podocytes/ultrastructure , Protein Transport/drug effects , RNA Interference , Silver Nitrate/toxicity , Survival Analysis , Tretinoin/metabolism
9.
Front Neurol ; 9: 473, 2018.
Article in English | MEDLINE | ID: mdl-30061855

ABSTRACT

After respiratory distress, cardiac dysfunction is the second most common cause of fatality associated with the myotonic dystrophy (DM) disease. Despite the prevalance of heart failure in DM, physiopathological studies on heart symptoms have been relatively scarce because few murine models faithfully reproduce the cardiac disease. Consequently, only a small number of candidate compounds have been evaluated in this specific phenotype. To help cover this gap Drosophila combines the amenability of its invertebrate genetics with the possibility of quickly acquiring physiological parameters suitable for meaningful comparisons with vertebrate animal models and humans. Here we review available descriptions of cardiac disease in DM type 1 and type 2, and three recent papers reporting the cardiac toxicity of non-coding CUG (DM1) and CCUG (DM2) repeat RNA in flies. Notably, flies expressing CUG or CCUG RNA in their hearts developed strong arrhythmias and had reduced fractional shortening, which correlates with similar phenotypes in DM patients. Overexpression of Muscleblind, which is abnormally sequestered by CUG and CCUG repeat RNA, managed to strongly suppress arrhythmias and fractional shortening, thus demonstrating that Muscleblind depletion causes cardiac phenotypes in flies. Importantly, small molecules pentamidine and daunorubicin were able to rescue cardiac phenotypes by releasing Muscleblind from sequestration. Taken together, fly heart models have the potential to make important contributions to the understanding of the molecular causes of cardiac dysfunction in DM and in the quick assessment of candidate therapeutics.

10.
Drug Discov Today ; 23(12): 2013-2022, 2018 12.
Article in English | MEDLINE | ID: mdl-30086404

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.


Subject(s)
Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , RNA/genetics , Animals , Humans , Myotonin-Protein Kinase/genetics , RNA, Messenger/genetics
11.
Nat Commun ; 9(1): 2482, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29946070

ABSTRACT

Functional depletion of the alternative splicing factors Muscleblind-like (MBNL 1 and 2) is at the basis of the neuromuscular disease myotonic dystrophy type 1 (DM1). We previously showed the efficacy of miRNA downregulation in Drosophila DM1 model. Here, we screen for miRNAs that regulate MBNL1 and MBNL2 in HeLa cells. We thus identify miR-23b and miR-218, and confirm that they downregulate MBNL proteins in this cell line. Antagonists of miR-23b and miR-218 miRNAs enhance MBNL protein levels and rescue pathogenic missplicing events in DM1 myoblasts. Systemic delivery of these "antagomiRs" similarly boost MBNL expression and improve DM1-like phenotypes, including splicing alterations, histopathology, and myotonia in the HSALR DM1 model mice. These mammalian data provide evidence for therapeutic blocking of the miRNAs that control Muscleblind-like protein expression in myotonic dystrophy.


Subject(s)
MicroRNAs/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , 3' Untranslated Regions , Alternative Splicing , Animals , Cell Line , Disease Models, Animal , Gene Silencing , HeLa Cells , Humans , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Myotonic Dystrophy/physiopathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
12.
Nat Commun ; 9(1): 2009, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29789616

ABSTRACT

Myotonic dystrophy type 1 and type 2 (DM1, DM2) are caused by expansions of CTG and CCTG repeats, respectively. RNAs containing expanded CUG or CCUG repeats interfere with the metabolism of other RNAs through titration of the Muscleblind-like (MBNL) RNA binding proteins. DM2 follows a more favorable clinical course than DM1, suggesting that specific modifiers may modulate DM severity. Here, we report that the rbFOX1 RNA binding protein binds to expanded CCUG RNA repeats, but not to expanded CUG RNA repeats. Interestingly, rbFOX1 competes with MBNL1 for binding to CCUG expanded repeats and overexpression of rbFOX1 partly releases MBNL1 from sequestration within CCUG RNA foci in DM2 muscle cells. Furthermore, expression of rbFOX1 corrects alternative splicing alterations and rescues muscle atrophy, climbing and flying defects caused by expression of expanded CCUG repeats in a Drosophila model of DM2.


Subject(s)
Muscle, Skeletal/metabolism , Myotonic Dystrophy/genetics , RNA Splicing Factors/chemistry , RNA-Binding Proteins/chemistry , RNA/chemistry , Animals , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Models, Molecular , Muscle, Skeletal/pathology , Myotonic Dystrophy/classification , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Nucleotide Motifs , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA/genetics , RNA/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
13.
J Vis Exp ; (133)2018 03 31.
Article in English | MEDLINE | ID: mdl-29658931

ABSTRACT

Muscle mass wasting, known as muscle atrophy, is a common phenotype in Drosophila models of neuromuscular diseases. We have used the indirect flight muscles (IFMs) of flies, specifically the dorso-longitudinal muscles (DLM), as the experimental subject to measure the atrophic phenotype brought about by different genetic causes. In this protocol, we describe how to embed fly thorax muscles for semi thin sectioning, how to obtain a good contrast between muscle and the surrounding tissue, and how to process optical microscope images for semiautomatic acquisition of quantifiable data and analysis. We describe three specific applications of the methodological pipeline. First, we show how the method can be applied to quantify muscle degeneration in a myotonic dystrophy fly model; second, measurement of muscle cross-sectional area can help to identify genes that either promote or prevent muscle atrophy and/or muscle degeneration; third, this protocol can be applied to determine whether a candidate compound is able to significantly modify a given atrophic phenotype induced by a disease-causing mutation or by an environmental trigger.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Muscles/surgery , Animals , Cross-Sectional Studies , Muscles/pathology
14.
Dis Model Mech ; 11(4)2018 04 23.
Article in English | MEDLINE | ID: mdl-29592894

ABSTRACT

Myotonic dystrophy (DM) is a dominantly inherited neuromuscular disorder caused by expression of mutant myotonin-protein kinase (DMPK) transcripts containing expanded CUG repeats. Pathogenic DMPK RNA sequesters the muscleblind-like (MBNL) proteins, causing alterations in metabolism of various RNAs. Cardiac dysfunction represents the second most common cause of death in DM type 1 (DM1) patients. However, the contribution of MBNL sequestration in DM1 cardiac dysfunction is unclear. We overexpressed Muscleblind (Mbl), the DrosophilaMBNL orthologue, in cardiomyocytes of DM1 model flies and observed a rescue of heart dysfunctions, which are characteristic of these model flies and resemble cardiac defects observed in patients. We also identified a drug - daunorubicin hydrochloride - that directly binds to CUG repeats and alleviates Mbl sequestration in Drosophila DM1 cardiomyocytes, resulting in mis-splicing rescue and cardiac function recovery. These results demonstrate the relevance of Mbl sequestration caused by expanded-CUG-repeat RNA in cardiac dysfunctions in DM1, and highlight the potential of strategies aimed at inhibiting this protein-RNA interaction to recover normal cardiac function.


Subject(s)
Daunorubicin/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Heart/physiopathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Alternative Splicing/genetics , Animals , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Heart/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Protein Binding/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Survival Analysis
15.
Dis Model Mech ; 11(1)2018 01 17.
Article in English | MEDLINE | ID: mdl-29208631

ABSTRACT

Members of the Junctophilin (JPH) protein family have emerged as key actors in all excitable cells, with crucial implications for human pathophysiology. In mammals, this family consists of four members (JPH1-JPH4) that are differentially expressed throughout excitable cells. The analysis of knockout mice lacking JPH subtypes has demonstrated their essential contribution to physiological functions in skeletal and cardiac muscles and in neurons. Moreover, mutations in the human JPH2 gene are associated with hypertrophic and dilated cardiomyopathies; mutations in JPH3 are responsible for the neurodegenerative Huntington's disease-like-2 (HDL2), whereas JPH1 acts as a genetic modifier in Charcot-Marie-Tooth 2K peripheral neuropathy. Drosophila melanogaster has a single junctophilin (jp) gene, as is the case in all invertebrates, which might retain equivalent functions of the four homologous JPH genes present in mammalian genomes. Therefore, owing to the lack of putatively redundant genes, a jpDrosophila model could provide an excellent platform to model the Junctophilin-related diseases, to discover the ancestral functions of the JPH proteins and to reveal new pathways. By up- and downregulation of Jp in a tissue-specific manner in Drosophila, we show that altering its levels of expression produces a phenotypic spectrum characterized by muscular deficits, dilated cardiomyopathy and neuronal alterations. Importantly, our study has demonstrated that Jp modifies the neuronal degeneration in a Drosophila model of Huntington's disease, and it has allowed us to uncover an unsuspected functional relationship with the Notch pathway. Therefore, this Drosophila model has revealed new aspects of Junctophilin function that can be relevant for the disease mechanisms of their human counterparts.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Huntingtin Protein/metabolism , Mammals/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Trinucleotide Repeat Expansion/genetics , Alleles , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster/ultrastructure , Gene Knockdown Techniques , Genes, Insect , Genotype , Muscles/metabolism , Muscles/pathology , Muscles/ultrastructure , Mutation/genetics , Myocardium/metabolism , Myocardium/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Phenotype , Photoreceptor Cells, Invertebrate/metabolism , RNA Interference , Reproducibility of Results
16.
Sci Rep ; 7(1): 2843, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28588248

ABSTRACT

Myotonic dystrophies (DM1-2) are neuromuscular genetic disorders caused by the pathological expansion of untranslated microsatellites. DM1 and DM2, are caused by expanded CTG repeats in the 3'UTR of the DMPK gene and CCTG repeats in the first intron of the CNBP gene, respectively. Mutant RNAs containing expanded repeats are retained in the cell nucleus, where they sequester nuclear factors and cause alterations in RNA metabolism. However, for unknown reasons, DM1 is more severe than DM2. To study the differences and similarities in the pathogenesis of DM1 and DM2, we generated model flies by expressing pure expanded CUG ([250]×) or CCUG ([1100]×) repeats, respectively, and compared them with control flies expressing either 20 repeat units or GFP. We observed surprisingly severe muscle reduction and cardiac dysfunction in CCUG-expressing model flies. The muscle and cardiac tissue of both DM1 and DM2 model flies showed DM1-like phenotypes including overexpression of autophagy-related genes, RNA mis-splicing and repeat RNA aggregation in ribonuclear foci along with the Muscleblind protein. These data reveal, for the first time, that expanded non-coding CCUG repeat-RNA has similar in vivo toxicity potential as expanded CUG RNA in muscle and heart tissues and suggests that specific, as yet unknown factors, quench CCUG-repeat toxicity in DM2 patients.


Subject(s)
DNA Repeat Expansion , Myotonic Dystrophy/genetics , Myotonin-Protein Kinase/genetics , Animals , Arrhythmias, Cardiac/etiology , Autophagy/genetics , Disease Models, Animal , Drosophila , Gene Expression , Locomotion , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myotonic Dystrophy/mortality , Myotonic Dystrophy/physiopathology , RNA Splicing
17.
PLoS One ; 12(6): e0178931, 2017.
Article in English | MEDLINE | ID: mdl-28582438

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a rare multisystemic disorder associated with an expansion of CUG repeats in mutant DMPK (dystrophia myotonica protein kinase) transcripts; the main effect of these expansions is the induction of pre-mRNA splicing defects by sequestering muscleblind-like family proteins (e.g. MBNL1). Disruption of the CUG repeats and the MBNL1 protein complex has been established as the best therapeutic approach for DM1, hence two main strategies have been proposed: targeted degradation of mutant DMPK transcripts and the development of CUG-binding molecules that prevent MBNL1 sequestration. Herein, suitable CUG-binding small molecules were selected using in silico approaches such as scaffold analysis, similarity searching, and druggability analysis. We used polarization assays to confirm the CUG repeat binding in vitro for a number of candidate compounds, and went on to evaluate the biological activity of the two with the strongest affinity for CUG repeats (which we refer to as compounds 1-2 and 2-5) in DM1 mutant cells and Drosophila DM1 models with an impaired locomotion phenotype. In particular, 1-2 and 2-5 enhanced the levels of free MBNL1 in patient-derived myoblasts in vitro and greatly improved DM1 fly locomotion in climbing assays. This work provides new computational approaches for rational large-scale virtual screens of molecules that selectively recognize CUG structures. Moreover, it contributes valuable knowledge regarding two compounds with desirable biological activity in DM1 models.


Subject(s)
Anabolic Agents/pharmacology , Benzamidines/pharmacology , Drosophila Proteins/antagonists & inhibitors , Myotonic Dystrophy/drug therapy , Myotonin-Protein Kinase/antagonists & inhibitors , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Alternative Splicing , Anabolic Agents/chemistry , Animals , Benzamidines/chemistry , Disease Models, Animal , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Drug Discovery , Humans , Locomotion/drug effects , Molecular Docking Simulation , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/chemistry , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Primary Cell Culture , Pyrimidines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Trinucleotide Repeat Expansion/drug effects
18.
Sci Rep ; 6: 36230, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27805016

ABSTRACT

Myotonic Dystrophy type 1 (DM1) originates from alleles of the DMPK gene with hundreds of extra CTG repeats in the 3' untranslated region (3' UTR). CUG repeat RNAs accumulate in foci that sequester Muscleblind-like (MBNL) proteins away from their functional target transcripts. Endogenous upregulation of MBNL proteins is, thus, a potential therapeutic approach to DM1. Here we identify two miRNAs, dme-miR-277 and dme-miR-304, that differentially regulate muscleblind RNA isoforms in miRNA sensor constructs. We also show that their sequestration by sponge constructs derepresses endogenous muscleblind not only in a wild type background but also in a DM1 Drosophila model expressing non-coding CUG trinucleotide repeats throughout the musculature. Enhanced muscleblind expression resulted in significant rescue of pathological phenotypes, including reversal of several mis-splicing events and reduced muscle atrophy in DM1 adult flies. Rescued flies had improved muscle function in climbing and flight assays, and had longer lifespan compared to disease controls. These studies provide proof of concept for a similar potentially therapeutic approach to DM1 in humans.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , MicroRNAs/genetics , Myotonic Dystrophy/genetics , Nuclear Proteins/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Flight, Animal/physiology , Gene Expression Regulation , Motor Activity/genetics , Motor Activity/physiology , Myotonic Dystrophy/metabolism , Nuclear Proteins/metabolism , Phenotype , Trinucleotide Repeat Expansion/genetics
19.
PLoS One ; 11(2): e0150501, 2016.
Article in English | MEDLINE | ID: mdl-26919350

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an autosomal dominant genetic disease caused by expansion of a CTG microsatellite in the 3' untranslated region of the DMPK gene. Despite characteristic muscular, cardiac, and neuropsychological symptoms, CTG trinucleotide repeats are unstable both in the somatic and germinal lines, making the age of onset, clinical presentation, and disease severity very variable. A molecular biomarker to stratify patients and to follow disease progression is, thus, an unmet medical need. Looking for a novel biomarker, and given that specific miRNAs have been found to be misregulated in DM1 heart and muscle tissues, we profiled the expression of 175 known serum miRNAs in DM1 samples. The differences detected between patients and controls were less than 2.6 fold for all of them and a selection of six candidate miRNAs, miR-103, miR-107, miR-21, miR-29a, miR-30c, and miR-652 all failed to show consistent differences in serum expression in subsequent validation experiments.


Subject(s)
MicroRNAs/blood , Myotonic Dystrophy/blood , Adult , Biomarkers , Blotting, Southern , Gene Expression Profiling , Humans , Male , Middle Aged , Myotonic Dystrophy/genetics , Trinucleotide Repeats
20.
Dis Model Mech ; 8(12): 1569-78, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26515653

ABSTRACT

Up to 80% of individuals with myotonic dystrophy type 1 (DM1) will develop cardiac abnormalities at some point during the progression of their disease, the most common of which is heart blockage of varying degrees. Such blockage is characterized by conduction defects and supraventricular and ventricular tachycardia, and carries a high risk of sudden cardiac death. Despite its importance, very few animal model studies have focused on the heart dysfunction in DM1. Here, we describe the characterization of the heart phenotype in a Drosophila model expressing pure expanded CUG repeats under the control of the cardiomyocyte-specific driver GMH5-Gal4. Morphologically, expression of 250 CUG repeats caused abnormalities in the parallel alignment of the spiral myofibrils in dissected fly hearts, as revealed by phalloidin staining. Moreover, combined immunofluorescence and in situ hybridization of Muscleblind and CUG repeats, respectively, confirmed detectable ribonuclear foci and Muscleblind sequestration, characteristic features of DM1, exclusively in flies expressing the expanded CTG repeats. Similarly to what has been reported in humans with DM1, heart-specific expression of toxic RNA resulted in reduced survival, increased arrhythmia, altered diastolic and systolic function, reduced heart tube diameters and reduced contractility in the model flies. As a proof of concept that the fly heart model can be used for in vivo testing of promising therapeutic compounds, we fed flies with pentamidine, a compound previously described to improve DM1 phenotypes. Pentamidine not only released Muscleblind from the CUG RNA repeats and reduced ribonuclear formation in the Drosophila heart, but also rescued heart arrhythmicity and contractility, and improved fly survival in animals expressing 250 CUG repeats.


Subject(s)
Drosophila melanogaster/physiology , Heart Rate/drug effects , Heart/physiopathology , Myocardial Contraction/drug effects , Myotonic Dystrophy/physiopathology , Pentamidine/pharmacology , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Diastole/drug effects , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Heart/drug effects , Longevity/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myotonic Dystrophy/pathology , Nuclear Proteins/metabolism , Phenotype , Survival Analysis , Systole/drug effects , Trinucleotide Repeat Expansion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...