Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nat Commun ; 12(1): 7299, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911927

ABSTRACT

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA Splicing , Small Molecule Libraries/administration & dosage , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Humans , Huntington Disease/metabolism , Mice , RNA Splicing/drug effects , RNA Stability/drug effects , Trinucleotide Repeat Expansion/drug effects
2.
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
3.
J Huntingtons Dis ; 10(1): 203-220, 2021.
Article in English | MEDLINE | ID: mdl-32925081

ABSTRACT

DNA damage repair (DDR) mechanisms have been implicated in a number of neurodegenerative diseases (both genetically determined and sporadic). Consistent with this, recent genome-wide association studies in Huntington's disease (HD) and other trinucleotide repeat expansion diseases have highlighted genes involved in DDR mechanisms as modifiers for age of onset, rate of progression and somatic instability. At least some clinical genetic modifiers have been shown to have a role in modulating trinucleotide repeat expansion biology and could therefore provide new disease-modifying therapeutic targets. In this review, we focus on key considerations with respect to drug discovery and development using DDR mechanisms as a target for trinucleotide repeat expansion diseases. Six areas are covered with specific reference to DDR and HD: 1) Target identification and validation; 2) Candidate selection including therapeutic modality and delivery; 3) Target drug exposure with particular focus on blood-brain barrier penetration, engagement and expression of pharmacology; 4) Safety; 5) Preclinical models as predictors of therapeutic efficacy; 6) Clinical outcome measures including biomarkers.


Subject(s)
DNA Damage/genetics , DNA Mismatch Repair/genetics , Drug Development , Drug Discovery , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Huntington Disease/genetics , Trinucleotide Repeat Expansion/genetics , Animals , DNA Damage/drug effects , DNA Mismatch Repair/drug effects , Humans , Huntingtin Protein/drug effects , Trinucleotide Repeat Expansion/drug effects
4.
Cell Chem Biol ; 28(1): 34-45.e6, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33157036

ABSTRACT

Many diseases are caused by toxic RNA repeats. Herein, we designed a lead small molecule that binds the structure of the r(CUG) repeat expansion [r(CUG)exp] that causes myotonic dystrophy type 1 (DM1) and Fuchs endothelial corneal dystrophy (FECD) and rescues disease biology in patient-derived cells and in vivo. Interestingly, the compound's downstream effects are different in the two diseases, owing to the location of the repeat expansion. In DM1, r(CUG)exp is harbored in the 3' untranslated region, and the compound has no effect on the mRNA's abundance. In FECD, however, r(CUG)exp is located in an intron, and the small molecule facilitates excision of the intron, which is then degraded by the RNA exosome complex. Thus, structure-specific, RNA-targeting small molecules can act disease specifically to affect biology, either by disabling the gain-of-function mechanism (DM1) or by stimulating quality control pathways to rid a disease-affected cell of a toxic RNA (FECD).


Subject(s)
Exosomes/drug effects , Fuchs' Endothelial Dystrophy/drug therapy , Myotonic Dystrophy/drug therapy , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cells, Cultured , Exosomes/metabolism , Female , Fuchs' Endothelial Dystrophy/metabolism , Humans , Male , Myotonic Dystrophy/metabolism , Trinucleotide Repeat Expansion/genetics
5.
Cell Mol Life Sci ; 78(4): 1577-1596, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32696070

ABSTRACT

Polyglutamine (polyQ) diseases are incurable neurological disorders caused by CAG repeat expansion in the open reading frames (ORFs) of specific genes. This type of mutation in the HTT gene is responsible for Huntington's disease (HD). CAG repeat-targeting artificial miRNAs (art-miRNAs) were shown as attractive therapeutic approach for polyQ disorders as they caused allele-selective decrease in the level of mutant proteins. Here, using polyQ disease models, we aimed to demonstrate how miRNA-based gene expression regulation is dependent on target sequence features. We show that the silencing efficiency and selectivity of art-miRNAs is influenced by the localization of the CAG repeat tract within transcript and the specific sequence context. Furthermore, we aimed to reveal the events leading to downregulation of mutant polyQ proteins and found very rapid activation of translational repression and HTT transcript deadenylation. Slicer-activity of AGO2 was dispensable in this process, as determined in AGO2 knockout cells generated with CRISPR-Cas9 technology. We also showed highly allele-selective downregulation of huntingtin in human HD neural progenitors (NPs). Taken together, art-miRNA activity may serve as a model of the cooperative activity and targeting of ORF regions by endogenous miRNAs.


Subject(s)
Argonaute Proteins/genetics , Huntingtin Protein/genetics , Huntington Disease/therapy , MicroRNAs/genetics , Alleles , CRISPR-Cas Systems/genetics , Gene Knockout Techniques , Humans , Huntington Disease/genetics , Huntington Disease/pathology , MicroRNAs/chemical synthesis , MicroRNAs/pharmacology , Mutation/genetics , Open Reading Frames/genetics , Peptides/genetics , Protein Biosynthesis/drug effects , RNA Interference , Trinucleotide Repeat Expansion/drug effects , Trinucleotide Repeat Expansion/genetics
6.
Biochemistry ; 59(37): 3463-3472, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32856901

ABSTRACT

There are few methods available for the rapid discovery of multitarget drugs. Herein, we describe the template-assisted, target-guided discovery of small molecules that recognize d(CTG) in the expanded d(CTG·CAG) sequence and its r(CUG) transcript that cause myotonic dystrophy type 1. A positive cross-selection was performed using a small library of 30 monomeric alkyne- and azide-containing ligands capable of producing >5000 possible di- and trimeric click products. The monomers were incubated with d(CTG)16 or r(CUG)16 under physiological conditions, and both sequences showed selectivity in the proximity-accelerated azide-alkyne [3+2] cycloaddition click reaction. The limited number of click products formed in both selections and the even smaller number of common products suggests that this method is a useful tool for the discovery of single-target and multitarget lead therapeutic agents.


Subject(s)
DNA/antagonists & inhibitors , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , RNA/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cells, Cultured , DNA/genetics , DNA/metabolism , Humans , Myotonic Dystrophy/pathology , RNA/genetics , RNA/metabolism , Trinucleotide Repeat Expansion/genetics
7.
ACS Chem Biol ; 15(4): 849-855, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32186845

ABSTRACT

RNA repeat expansions cause more than 30 neurological and neuromuscular diseases with no known cures. Since repeat expansions operate via diverse pathomechanisms, one potential therapeutic strategy is to rid them from disease-affected cells, using bifunctional small molecules that cleave the aberrant RNA. Such an approach has been previously implemented for the RNA repeat that causes myotonic dystrophy type 1 [DM1, r(CUG)exp] with Cugamycin, which is a small molecule that selectively binds r(CUG)exp conjugated to a bleomycin A5 cleaving module. Herein, we demonstrate that, by replacing bleomycin A5 with deglycobleomycin, an analogue in which the carbohydrate domain of bleomycin A5 is removed, the selectivity of the resulting small-molecule conjugate (DeglycoCugamycin) was enhanced, while maintaining potent and allele-selective cleavage of r(CUG)exp and rescue of DM1-associated defects. In particular, DeglycoCugamycin did not induce the DNA damage that is observed with high concentrations (25 µM) of Cugamycin, while selectively cleaving the disease-causing allele and improving DM1 defects at 1 µM.


Subject(s)
Bleomycin/analogs & derivatives , DNA Cleavage/drug effects , DNA/chemistry , Trinucleotide Repeat Expansion/drug effects , Animals , Bleomycin/chemistry , Bleomycin/pharmacology , Cell Line , DNA Damage/drug effects , DNA-Binding Proteins/genetics , Mice , RNA-Binding Proteins/genetics
8.
Nat Genet ; 52(2): 146-159, 2020 02.
Article in English | MEDLINE | ID: mdl-32060489

ABSTRACT

In many repeat diseases, such as Huntington's disease (HD), ongoing repeat expansions in affected tissues contribute to disease onset, progression and severity. Inducing contractions of expanded repeats by exogenous agents is not yet possible. Traditional approaches would target proteins driving repeat mutations. Here we report a compound, naphthyridine-azaquinolone (NA), that specifically binds slipped-CAG DNA intermediates of expansion mutations, a previously unsuspected target. NA efficiently induces repeat contractions in HD patient cells as well as en masse contractions in medium spiny neurons of HD mouse striatum. Contractions are specific for the expanded allele, independently of DNA replication, require transcription across the coding CTG strand and arise by blocking repair of CAG slip-outs. NA-induced contractions depend on active expansions driven by MutSß. NA injections in HD mouse striatum reduce mutant HTT protein aggregates, a biomarker of HD pathogenesis and severity. Repeat-structure-specific DNA ligands are a novel avenue to contract expanded repeats.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Naphthyridines/pharmacology , Quinolones/pharmacology , Trinucleotide Repeat Expansion/drug effects , Animals , Corpus Striatum/drug effects , DNA/metabolism , DNA Mismatch Repair/drug effects , DNA Replication/drug effects , Disease Models, Animal , Humans , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/pathology , Male , Mice , Mice, Transgenic , Microsatellite Instability , Mutation , Ribonucleases/metabolism , TATA-Box Binding Protein/genetics , Transcription, Genetic
9.
Nucleic Acid Ther ; 30(2): 80-93, 2020 04.
Article in English | MEDLINE | ID: mdl-31873063

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a dominantly inherited, multisystemic disorder characterized clinically by delayed muscle relaxation and weakness. The disease is caused by a CTG repeat expansion in the 3' untranslated region (3' UTR) of the DMPK gene, which leads to the expression of a toxic gain-of-function mRNA. The expanded CUG repeat mRNA sequesters the MBNL1 splicing regulator in nuclear-retained foci structures, resulting in loss of protein function and disruption of alternative splicing homeostasis. In this study, we used CAG repeat antisense oligonucleotides (ASOs), composed of locked nucleic acid (LNA)- and 2'-O-methyl (2'OMe)-modified bases in a chimeric design, to alleviate CUGexpanded-mediated toxicity. Chimeric 14-18mer LNA/2'OMe oligonucleotides, exhibiting an LNA incorporation of ∼33%, significantly ameliorated the misregulated alternative splicing of Mbnl1-dependent exons in primary DM1 mouse myoblasts and tibialis anterior muscles of DM1 mice. Subcutaneous delivery of 14mer and 18mer LNA/2'OMe chimeras in DM1 mice resulted in high levels of accumulation in all tested skeletal muscles, as well as in the diaphragm and heart tissue. Despite the efficient delivery, chimeric LNA/2'OMe oligonucleotides were not able, even at a high-dosage regimen (400 mg/kg/week), to correct the misregulated splicing of Serca1 exon 22 in skeletal muscles. Nevertheless, oligonucleotide doses were well-tolerated as determined by histological and plasma biochemistry analyses. Our results provide proof of concept that inhibition of MBNL1 sequestration by systemic delivery of a steric-blocking ASO is extremely challenging, considering the large number of target sites that need to be occupied per RNA molecule. Although not suitable for DM1 therapy, chimeric LNA/2'OMe oligonucleotides could prove to be highly beneficial for other diseases, such as Duchenne muscular dystrophy, that require inhibition of a single target site per RNA molecule.


Subject(s)
Alternative Splicing/drug effects , Myotonic Dystrophy/therapy , Myotonin-Protein Kinase/genetics , Trinucleotide Repeat Expansion/drug effects , 3' Untranslated Regions/genetics , Alternative Splicing/genetics , Animals , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Disease Models, Animal , Exons/genetics , Humans , Mice , Myotonic Dystrophy/genetics , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/antagonists & inhibitors , Oligonucleotides/genetics , Oligonucleotides/pharmacology , RNA Splicing/drug effects , RNA Splicing/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Trinucleotide Repeat Expansion/genetics
10.
Sci Rep ; 9(1): 16872, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31728006

ABSTRACT

CAG repeats RNA causes various fatal neurodegenerative diseases exemplified by Huntington's disease (HD) and several spinocerebellar ataxias (SCAs). Although there are differences in the pathogenic mechanisms, these diseases share the common cause, i.e., expansion of CAG repeats. The shared cause of these diseases raises the possibility for the exploiting the common target as a potential therapeutic approach. Oligonucleotide-based therapeutics are designed earlier with the help of the base pairing rule but are not very promiscuous, considering the nonspecific stimulation of the immune system and the poor cellular delivery. Therefore, small molecules-based therapeutics are preferred for targeting the repeats expansion disorders. Here, we have used the chemical similarity search approach to discern the small molecules that selectively target toxic CAG RNA. The lead compounds showed the specificity towards AA mismatch in biophysical studies including CD, ITC, and NMR spectroscopy and thus aided to forestall the polyQ mediated pathogenicity. Furthermore, the lead compounds also explicitly alleviate the polyQ mediated toxicity in HD cell models and patient-derived cells. These findings suggest that the lead compound could act as a chemical probe for AA mismatch containing RNA as well as plays a neuroprotective role in fatal neurodegenerative diseases like HD and SCAs.


Subject(s)
Fibroblasts/drug effects , Flavonoids/pharmacology , Neuroprotective Agents/pharmacology , Peptides/antagonists & inhibitors , RNA/chemistry , Small Molecule Libraries/pharmacology , Benzothiazoles/chemistry , Biological Assay , Cell Survival/drug effects , Drug Discovery , Fibroblasts/metabolism , Fibroblasts/pathology , Flavonoids/chemistry , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Molecular Docking Simulation , Neuroprotective Agents/chemistry , Nucleic Acid Conformation , Peptides/chemistry , Peptides/metabolism , Primary Cell Culture , Protein Aggregates/drug effects , RNA/genetics , RNA/metabolism , Small Molecule Libraries/chemistry , Trinucleotide Repeat Expansion/drug effects
11.
J Biol Chem ; 294(49): 18624-18638, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31649034

ABSTRACT

Repeat-associated non-AUG (RAN) translation is a noncanonical translation initiation event that occurs at nucleotide-repeat expansion mutations that are associated with several neurodegenerative diseases, including fragile X-associated tremor ataxia syndrome (FXTAS), ALS, and frontotemporal dementia (FTD). Translation of expanded repeats produces toxic proteins that accumulate in human brains and contribute to disease pathogenesis. Consequently, RAN translation constitutes a potentially important therapeutic target for managing multiple neurodegenerative disorders. Here, we adapted a previously developed RAN translation assay to a high-throughput format to screen 3,253 bioactive compounds for inhibition of RAN translation of expanded CGG repeats associated with FXTAS. We identified five diverse small molecules that dose-dependently inhibited CGG RAN translation, while relatively sparing canonical translation. All five compounds also inhibited RAN translation of expanded GGGGCC repeats associated with ALS and FTD. Using CD and native gel analyses, we found evidence that three of these compounds, BIX01294, CP-31398, and propidium iodide, bind directly to the repeat RNAs. These findings provide proof-of-principle supporting the development of selective small-molecule RAN translation inhibitors that act across multiple disease-causing repeats.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Ataxia/genetics , Fragile X Syndrome/genetics , Tremor/genetics , Trinucleotide Repeat Expansion/genetics , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Ataxia/drug therapy , Azepines/pharmacology , Azepines/therapeutic use , Cells, Cultured , Circular Dichroism , DNA Repeat Expansion/drug effects , DNA Repeat Expansion/genetics , Drug Evaluation, Preclinical , Fragile X Syndrome/drug therapy , HEK293 Cells , Humans , Neurodegenerative Diseases/genetics , Propidium/pharmacology , Propidium/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Rats , Tremor/drug therapy , Trinucleotide Repeat Expansion/drug effects
12.
Proc Natl Acad Sci U S A ; 116(42): 20991-21000, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570586

ABSTRACT

A CTG repeat expansion in the DMPK gene is the causative mutation of myotonic dystrophy type 1 (DM1). Transcription of the expanded CTG repeat produces toxic gain-of-function CUG RNA, leading to disease symptoms. A screening platform that targets production or stability of the toxic CUG RNA in a selective manner has the potential to provide new biological and therapeutic insights. A DM1 HeLa cell model was generated that stably expresses a toxic r(CUG)480 and an analogous r(CUG)0 control from DMPK and was used to measure the ratio-metric level of r(CUG)480 versus r(CUG)0. This DM1 HeLa model recapitulates pathogenic hallmarks of DM1, including CUG ribonuclear foci and missplicing of pre-mRNA targets of the muscleblind (MBNL) alternative splicing factors. Repeat-selective screening using this cell line led to the unexpected identification of multiple microtubule inhibitors as hits that selectively reduce r(CUG)480 levels and partially rescue MBNL-dependent missplicing. These results were validated by using the Food and Drug Administration-approved clinical microtubule inhibitor colchicine in DM1 mouse and primary patient cell models. The mechanism of action was found to involve selective reduced transcription of the CTG expansion that we hypothesize to involve the LINC (linker of nucleoskeleton and cytoskeleton) complex. The unanticipated identification of microtubule inhibitors as selective modulators of toxic CUG RNA opens research directions for this form of muscular dystrophy and may shed light on the biology of CTG repeat expansion and inform therapeutic avenues. This approach has the potential to identify modulators of expanded repeat-containing gene expression for over 30 microsatellite expansion disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Microtubules/drug effects , Myotonic Dystrophy/genetics , RNA/genetics , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Animals , HeLa Cells , Humans , Mice , Mice, Transgenic , Microtubules/genetics , Microtubules/metabolism , Myotonic Dystrophy/enzymology , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , RNA/chemistry , RNA/metabolism
13.
ACS Chem Neurosci ; 10(8): 3778-3788, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31264835

ABSTRACT

An expansion of CGG tandem repeats in the 5' untranslated region (5'-UTR) of fragile X mental retardation 1 (FMR1) gene causes fragile X-associated tremor/ataxia syndrome (FXTAS). The transcripts of these expanded repeats r(CGG)exp either form RNA foci or undergo the repeat-associated non-ATG (RAN) translation that produces toxic homopolymeric proteins in neuronal cells. The discovery of small molecule modulators that possess a strong binding affinity and high selectivity to these toxic expanded repeats RNA could be a promising therapeutic approach to cure the expanded repeat-associated neurological diseases. Therefore, here we sought to test the therapeutic potential of a natural alkaloid, piperine, by assessing its ability to bind and neutralize the toxicity of r(CGG)exp RNA motif. To accomplish this first, we have determined the affinity of piperine to r(CGG)exp RNA using fluorescence-based binding assay and isothermal titration calorimetry assay. These assays showed that piperine forms a thermodynamically favorable interaction with r(CGG)exp RNA with high selectivity to the G-rich RNA motif. Interaction of piperine with r(CGG)exp motif was further validated using several biophysical techniques such as CD, CD melting, NMR spectroscopy, and gel retardation assay. Moreover, piperine was also found to be effective for improving the r(CGG)exp associated splicing defects and RAN translation in a FXTAS cell model system. Our results effectively provided the evidence that piperine strongly interacts with r(CGG)exp RNA and could be used as a suitable candidate for therapeutic development against FXTAS.


Subject(s)
Alkaloids/pharmacology , Ataxia/metabolism , Benzodioxoles/pharmacology , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Tremor/metabolism , Trinucleotide Repeat Expansion/drug effects , Humans
14.
Nucleic Acid Ther ; 29(2): 73-81, 2019 04.
Article in English | MEDLINE | ID: mdl-30676271

ABSTRACT

Fuchs' endothelial corneal dystrophy (FECD) leads to vision loss and is one of the most common inherited eye diseases. Corneal transplants are the only curative treatment available, and there is a major unmet need for treatments that are less invasive and independent of donor tissue. Most cases of FECD are associated with an expanded CUG repeat within the intronic region of TCF4 and the mutant RNA has been implicated as the cause of the disease. We previously presented preliminary data suggesting that single-stranded antisense oligonucleotides (ASOs) can inhibit CUG RNA foci in patient-derived cells and tissue. We now show that duplex RNAs and single-stranded silencing RNAs (ss-siRNAs) reduce the number of cells with foci and the number of foci per cells. Potencies are similar to those that are achieved with chemically modified ASOs designed to block foci. These data widen the potential for synthetic nucleic acids to be used to treat a widely prevalent and debilitating disease.


Subject(s)
Fuchs' Endothelial Dystrophy/genetics , Oligonucleotides, Antisense/pharmacology , RNA, Small Interfering/pharmacology , Cell Line , Fuchs' Endothelial Dystrophy/pathology , Fuchs' Endothelial Dystrophy/therapy , Gene Silencing/drug effects , Genetic Predisposition to Disease , Humans , Introns/drug effects , Oligonucleotides, Antisense/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/pharmacology , RNA, Small Interfering/genetics , Transcription Factor 4/antagonists & inhibitors , Transcription Factor 4/genetics , Trinucleotide Repeat Expansion/drug effects , Trinucleotide Repeat Expansion/genetics
15.
Bioorg Med Chem Lett ; 28(17): 2850-2855, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30076049

ABSTRACT

Friedreich's ataxia (FRDA) is an incurable neurodegenerative disorder caused by reduced expression of the mitochondrial protein frataxin (FXN). The genetic cause of the disease is an expanded GAA repeat within the FXN gene. Agents that increase expression of FXN protein are a potential approach to therapy. We previously described anti-trinucleotide GAA duplex RNAs (dsRNAs) and antisense oligonucleotides (ASOs) that activate FXN protein expression in multiple patient derived cell lines. Here we test two distinct series of compounds for their ability to increase FXN expression. ASOs with butane linkers showed low potency, which is consistent with the low Tm values and suggesting that flexible conformation impairs activity. By contrast, single-stranded siRNAs (ss-siRNAs) that combine the strengths of dsRNA and ASO approaches had nanomolar potencies. ss-siRNAs provide an additional option for developing nucleic acid therapeutics to treat FRDA.


Subject(s)
Friedreich Ataxia/drug therapy , Iron-Binding Proteins/genetics , RNA, Small Interfering/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cell Line , Dose-Response Relationship, Drug , Friedreich Ataxia/genetics , Humans , Iron-Binding Proteins/metabolism , Molecular Structure , Structure-Activity Relationship , Trinucleotide Repeat Expansion/genetics , Frataxin
16.
ACS Chem Neurosci ; 9(6): 1399-1408, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29506378

ABSTRACT

Expanded CAG trinucleotide repeats in Huntington's disease (HD) are causative for neurotoxicity. The mutant CAG repeat RNA encodes neurotoxic polyglutamine proteins and can lead to a toxic gain of function by aberrantly recruiting RNA-binding proteins. One of these is the MID1 protein, which induces aberrant Huntingtin (HTT) protein translation upon binding. Here we have identified a set of CAG repeat binder candidates by in silico methods. One of those, furamidine, reduces the level of binding of HTT mRNA to MID1 and other target proteins in vitro. Metadynamics calculations, fairly consistent with experimental data measured here, provide hints about the binding mode of the ligand. Importantly, furamidine also decreases the protein level of HTT in a HD cell line model. This shows that small molecules masking RNA-MID1 interactions may be active against mutant HTT protein in living cells.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Trinucleotide Repeat Expansion/drug effects , Cell Line/drug effects , Humans , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Nerve Tissue Proteins/genetics , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Peptides/pharmacology , RNA, Messenger/metabolism , Trinucleotide Repeat Expansion/genetics
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.
Nat Chem Biol ; 13(2): 188-193, 2017 02.
Article in English | MEDLINE | ID: mdl-27941760

ABSTRACT

Excluding the ribosome and riboswitches, developing small molecules that selectively target RNA is a longstanding problem in chemical biology. A typical cellular RNA is difficult to target because it has little tertiary, but abundant secondary structure. We designed allele-selective compounds that target such an RNA, the toxic noncoding repeat expansion (r(CUG)exp) that causes myotonic dystrophy type 1 (DM1). We developed several strategies to generate allele-selective small molecules, including non-covalent binding, covalent binding, cleavage and on-site probe synthesis. Covalent binding and cleavage enabled target profiling in cells derived from individuals with DM1, showing precise recognition of r(CUG)exp. In the on-site probe synthesis approach, small molecules bound adjacent sites in r(CUG)exp and reacted to afford picomolar inhibitors via a proximity-based click reaction only in DM1-affected cells. We expanded this approach to image r(CUG)exp in its natural context.


Subject(s)
RNA/chemistry , RNA/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Trinucleotide Repeat Expansion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Molecular Structure , RNA/genetics , RNA Splicing/drug effects , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship
19.
Neuron ; 90(5): 941-7, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27210553

ABSTRACT

Pathological evidence for selective four-repeat (4R) tau deposition in certain dementias and exon 10-positioned MAPT mutations together suggest a 4R-specific role in causing disease. However, direct assessments of 4R toxicity have not yet been accomplished in vivo. Increasing 4R-tau expression without change to total tau in human tau-expressing mice induced more severe seizures and nesting behavior abnormality, increased tau phosphorylation, and produced a shift toward oligomeric tau. Exon 10 skipping could also be accomplished in vivo, providing support for a 4R-tau targeted approach to target 4R-tau toxicity and, in cases of primary MAPT mutation, eliminate the disease-causing mutation.


Subject(s)
Models, Biological , Nesting Behavior , Seizures/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Brain/metabolism , Exons/genetics , Humans , Infusions, Intraventricular , Mice , Mutation/drug effects , Nesting Behavior/drug effects , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Protein Isoforms/metabolism , RNA Splicing/drug effects , RNA Splicing/genetics , Seizures/chemically induced , Seizures/genetics , Solubility , Trinucleotide Repeat Expansion/drug effects , tau Proteins/genetics , tau Proteins/toxicity
20.
Cell Rep ; 13(11): 2386-2394, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686629

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an inherited disease characterized by the inability to relax contracted muscles. Affected individuals carry large CTG expansions that are toxic when transcribed. One possible treatment approach is to reduce or eliminate transcription of CTG repeats. Actinomycin D (ActD) is a potent transcription inhibitor and FDA-approved chemotherapeutic that binds GC-rich DNA with high affinity. Here, we report that ActD decreased CUG transcript levels in a dose-dependent manner in DM1 cell and mouse models at significantly lower concentrations (nanomolar) compared to its use as a general transcription inhibitor or chemotherapeutic. ActD also significantly reversed DM1-associated splicing defects in a DM1 mouse model, and did so within the currently approved human treatment range. RNA-seq analyses showed that low concentrations of ActD did not globally inhibit transcription in a DM1 mouse model. These results indicate that transcription inhibition of CTG expansions is a promising treatment approach for DM1.


Subject(s)
Dactinomycin/pharmacology , Myotonic Dystrophy/pathology , RNA/metabolism , Trinucleotide Repeat Expansion/drug effects , Animals , Autophagy-Related Proteins , Base Sequence , Calorimetry , Chloride Channels/genetics , Chloride Channels/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , Mice , Microscopy, Fluorescence , Myotonic Dystrophy/metabolism , RNA/chemistry , RNA Splicing/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sequence Analysis, RNA , Transcription, Genetic/drug effects , Trinucleotide Repeat Expansion/genetics , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...