Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










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.
ChemMedChem ; 16(17): 2638-2644, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34114350

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a multisystemic neuromuscular disorder that is inherited in an autosomal dominant manner. DM1 originates in a (CTG⋅CAG) repeat expansion in the 3'-UTR of the dystrophia myotonic protein kinase (DMPK) gene on chromosome 19. One of the transcripts, r(CUG)exp , is toxic in various ways. Herein we report a rationally designed small molecule with a thiazole peptidomimetic unit that can serve as a minor groove binder for the nucleic acid targets. This peptide unit linked to two triaminotriazine recognition units selectively binds to d(CTG)exp to inhibit the transcription process, and also targets r(CUG)exp selectively to improve representative DM1 pathological molecular features, including foci formation and pre-mRNA splicing defects in DM1 model cells. As such, it represents a new structure type that might serve as a lead compound for future structure-activity optimization.


Subject(s)
Myotonic Dystrophy/drug therapy , Peptidomimetics/pharmacology , Small Molecule Libraries/pharmacology , Thiazoles/pharmacology , Triazines/pharmacology , Trinucleotide Repeats/drug effects , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Dynamics Simulation , Molecular Structure , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Peptidomimetics/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Thiazoles/chemistry , Triazines/chemistry , Trinucleotide Repeats/genetics
3.
Chem Commun (Camb) ; 57(26): 3235-3238, 2021 Apr 04.
Article in English | MEDLINE | ID: mdl-33646236

ABSTRACT

We demonstrated that a synthetic ligand NA, which selectively binds to a 5'-CAG-3'/5'-CAG-3' triad, induced repeat contractions during DNA polymerase-mediated primer extension through the CAG repeat template. A thorough capillary electrophoresis and sequencing analysis revealed that the d(CAG)20 template gave shortened nascent strands mainly containing 3-6 CTG units in the presence of NA.


Subject(s)
DNA/genetics , Naphthyridines/pharmacology , Quinolones/pharmacology , Trinucleotide Repeats/drug effects , DNA Replication/drug effects , Electrophoresis, Capillary , Humans , Ligands , Nucleic Acid Conformation/drug effects
5.
Int J Mol Sci ; 19(9)2018 Sep 17.
Article in English | MEDLINE | ID: mdl-30227633

ABSTRACT

We have reported the propensity of a DNA sequence containing CCG repeats to form a stable i-motif tetraplex structure in the absence of ligands. Here we show that an i-motif DNA sequence may transition to a base-extruded duplex structure with a GGCC tetranucleotide tract when bound to the (CoII)-mediated dimer of chromomycin A3, CoII(Chro)2. Biophysical experiments reveal that CCG trinucleotide repeats provide favorable binding sites for CoII(Chro)2. In addition, water hydration and divalent metal ion (CoII) interactions also play a crucial role in the stabilization of CCG trinucleotide repeats (TNRs). Our data furnish useful structural information for the design of novel therapeutic strategies to treat neurological diseases caused by repeat expansions.


Subject(s)
Chromomycin A3/pharmacology , Cobalt/pharmacology , Coordination Complexes/pharmacology , DNA/chemistry , Nucleic Acid Conformation/drug effects , Trinucleotide Repeats/drug effects , Chromomycin A3/chemistry , Cobalt/chemistry , Coordination Complexes/chemistry , Crystallography, X-Ray , Drug Discovery , Models, Molecular
6.
EMBO Rep ; 19(3)2018 03.
Article in English | MEDLINE | ID: mdl-29440125

ABSTRACT

Trinucleotide repeat (TNR) expansions in the genome cause a number of degenerative diseases. A prominent TNR expansion involves the triplet CAG in the huntingtin (HTT) gene responsible for Huntington's disease (HD). Pathology is caused by protein and RNA generated from the TNR regions including small siRNA-sized repeat fragments. An inverse correlation between the length of the repeats in HTT and cancer incidence has been reported for HD patients. We now show that siRNAs based on the CAG TNR are toxic to cancer cells by targeting genes that contain long reverse complementary TNRs in their open reading frames. Of the 60 siRNAs based on the different TNRs, the six members in the CAG/CUG family of related TNRs are the most toxic to both human and mouse cancer cells. siCAG/CUG TNR-based siRNAs induce cell death in vitro in all tested cancer cell lines and slow down tumor growth in a preclinical mouse model of ovarian cancer with no signs of toxicity to the mice. We propose to explore TNR-based siRNAs as a novel form of anticancer reagents.


Subject(s)
Huntingtin Protein/genetics , Neoplasms/genetics , RNA, Small Interfering/pharmacology , Trinucleotide Repeats/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Humans , Huntingtin Protein/antagonists & inhibitors , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Neoplasms/pathology , Neoplasms/therapy , Open Reading Frames , RNA, Small Interfering/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects
7.
RNA ; 24(4): 486-498, 2018 04.
Article in English | MEDLINE | ID: mdl-29295891

ABSTRACT

Polyglutamine (polyQ) diseases are a class of progressive neurodegenerative disorders characterized by the expression of both expanded CAG RNA and misfolded polyQ protein. We previously reported that the direct interaction between expanded CAG RNA and nucleolar protein nucleolin (NCL) impedes preribosomal RNA (pre-rRNA) transcription, and eventually triggers nucleolar stress-induced apoptosis in polyQ diseases. Here, we report that a 21-amino acid peptide, named "beta-structured inhibitor for neurodegenerative diseases" (BIND), effectively suppresses toxicity induced by expanded CAG RNA. When administered to a cell model, BIND potently inhibited cell death induced by expanded CAG RNA with an IC50 value of ∼0.7 µM. We showed that the function of BIND is dependent on Glu2, Lys13, Gly14, Ile18, Glu19, and Phe20. BIND treatment restored the subcellular localization of nucleolar marker protein and the expression level of pre-45s rRNA Through isothermal titration calorimetry analysis, we demonstrated that BIND suppresses nucleolar stress via a direct interaction with CAG RNA in a length-dependent manner. The mean binding constants (KD) of BIND to SCA2CAG22 , SCA2CAG42 , SCA2CAG55 , and SCA2CAG72 RNA are 17.28, 5.60, 4.83, and 0.66 µM, respectively. In vivo, BIND ameliorates retinal degeneration and climbing defects, and extends the lifespan of Drosophila expressing expanded CAG RNA. These effects suggested that BIND can suppress neurodegeneration in diverse polyQ disease models in vivo and in vitro without exerting observable cytotoxic effect. Our results collectively demonstrated that BIND is an effective inhibitor of expanded CAG RNA-induced toxicity in polyQ diseases.


Subject(s)
Huntington Disease/therapy , Peptides/pharmacology , Proteostasis Deficiencies/genetics , Spinocerebellar Ataxias/therapy , Trinucleotide Repeats/genetics , Animals , Cell Death/drug effects , Drosophila/genetics , HEK293 Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Peptides/metabolism , Phosphoproteins/genetics , Protein Folding , Proteostasis Deficiencies/pathology , Proteostasis Deficiencies/therapy , RNA, Ribosomal/genetics , RNA-Binding Proteins/genetics , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Transcription, Genetic/genetics , Trinucleotide Repeats/drug effects , Nucleolin
8.
Angew Chem Int Ed Engl ; 56(30): 8761-8765, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28544401

ABSTRACT

Small-molecule compounds targeting trinucleotide repeats in DNA have considerable potential as therapeutic or diagnostic agents against many neurological diseases. NiII (Chro)2 (Chro=chromomycin A3) binds specifically to the minor groove of (CCG)n repeats in duplex DNA, with unique fluorescence features that may serve as a probe for disease detection. Crystallographic studies revealed that the specificity originates from the large-scale spatial rearrangement of the DNA structure, including extrusion of consecutive bases and backbone distortions, with a sharp bending of the duplex accompanied by conformational changes in the NiII chelate itself. The DNA deformation of CCG repeats upon binding forms a GGCC tetranucleotide tract, which is recognized by NiII (Chro)2 . The extruded cytosine and last guanine nucleotides form water-mediated hydrogen bonds, which aid in ligand recognition. The recognition can be accounted for by the classic induced-fit paradigm.


Subject(s)
Chromomycins/pharmacology , DNA/drug effects , Nickel/pharmacology , Organometallic Compounds/pharmacology , Chromomycins/chemistry , DNA/chemistry , Humans , Models, Molecular , Nickel/chemistry , Organometallic Compounds/chemistry , Trinucleotide Repeats/drug effects
9.
Chemistry ; 22(42): 14881-14889, 2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27573860

ABSTRACT

The development of small molecules that can recognize specific RNA secondary and tertiary structures is currently an important research topic for developing tools to modulate gene expression and therapeutic drugs. Expanded CUG trinucleotide repeats, known as toxic RNA, capture the splicing factor MBNL1 and are causative of neurological disorder myotonic dystrophy type 1 (DM1). Herein, the rational molecular design, synthesis, and binding analysis of 2,9-diaminoalkyl-substituted 1,10-phenanthroline (DAP), which bound to CUG trinucleotide repeats, is described. The results of melting temperature (Tm ) analyses, surface plasmon resonance (SPR) assay, and electrospray spray ionization time-of-flight (ESI-TOF) mass spectrometry showed that DAP bound to r(CUG)9 but not to r(CAG)9 and r(CGG)9 . The dual luciferase assay clearly indicated DAP bound to the r(CUG)n repeat by affecting the translation in vitro.


Subject(s)
Myotonic Dystrophy/metabolism , Phenanthrolines/chemistry , RNA-Binding Proteins/metabolism , RNA/metabolism , Trinucleotide Repeat Expansion/physiology , Trinucleotide Repeats/drug effects , Humans , Ligands , RNA/chemistry , RNA Splicing , RNA-Binding Proteins/chemistry
10.
Bioorg Med Chem Lett ; 26(15): 3761-4, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27287365

ABSTRACT

We describe a new molecular design, synthesis, and investigation of small molecules that bind to CTG trinucleotide repeats in DNA. 1H-Pyrrolo[3,2-h]quinoline-8-amine (PQA) has a tricyclic aromatic system with unique non-linear hydrogen-bonding surface complementary to thymine. We have synthesized a series of PQA derivatives with different alkylamino linkers. These PQAs showed binding to pyrimidine bulge DNAs and CNG (N=T and C) repeats depending on the linker structure, while quinoline derivatives lacking the pyrrole ring showed much lower binding affinity. PQA is a useful molecular unit for both CTG and CCG repeat binding.


Subject(s)
Amines/pharmacology , DNA/drug effects , Pyrroles/pharmacology , Quinolines/pharmacology , Trinucleotide Repeats/drug effects , Amines/chemistry , Dose-Response Relationship, Drug , Ligands , Molecular Structure , Pyrroles/chemistry , Quinolines/chemistry , Structure-Activity Relationship
11.
Curr Top Med Chem ; 15(14): 1398-408, 2015.
Article in English | MEDLINE | ID: mdl-25866276

ABSTRACT

Expansion of trinucleotide repeats (TNRs) within genes plays a major role in pathology of various neurological diseases. The correlations of these unusual repetitive sequences with the aetiology of these diseases and the mechanism by which those repeats are expanded during replication have been extensively studied. Small-molecule ligands that bind to TNRs could provide potent biological applications. First, the length of the TNR is the most important determinant of these neurological diseases. Ligands that reduce the repeat length or impair repeat expansion may be used to delay onset and reduce the severity of these diseases. Interestingly, many important anticancer ligands and antibiotics have desirable qualities when interacting with TNR DNA, and may form the basis for the development of novel therapeutics against neurological diseases. Second, designed ligands that bind to expanded TNRs with high specificity based on the structural and chemical characteristics of these repeats can serve as diagnostic tools for determining repeat length and may have applications in preventive medicine. In this article we will review our current understanding of the interaction between DNA-binding ligands and TNRs.


Subject(s)
DNA/chemistry , DNA/drug effects , Nervous System Diseases/diagnosis , Nervous System Diseases/drug therapy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Trinucleotide Repeats/drug effects , Humans , Ligands , Nervous System Diseases/genetics , Trinucleotide Repeats/genetics
12.
Bioorg Med Chem ; 22(17): 4646-57, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25127467

ABSTRACT

Introducing novel building blocks to solid-phase peptide synthesis, we readily synthesized long-chain hairpin pyrrole-imidazole (PI) polyamide-chlorambucil conjugates 3 and 4 via the introduction of an amino group into a GABA (γ-turn) contained in 3, to target CAG/CTG repeat sequences, which are associated with various hereditary disorders. A high-resolution denaturing polyacrylamide sequencing gel revealed sequence-specific alkylation both strands at the N3 of adenines or guanines in CAG/CTG repeats by conjugates 3 and 4, with 11bp recognition. In vitro transcription assays using conjugate 4 revealed that specific alkylation inhibited the progression of RNA polymerase at the alkylating sites. Chiral substitution of the γ-turn with an amino group resulted in higher binding affinity observed in SPR assays. These assays suggest that conjugates 4 with 11bp recognition has the potential to cause specific DNA damage and transcriptional inhibition at the alkylating sites.


Subject(s)
Chlorambucil/pharmacology , DNA/drug effects , Imidazoles/pharmacology , Nylons/pharmacology , Pyrroles/pharmacology , Transcription, Genetic/drug effects , Trinucleotide Repeats/genetics , Alkylation/drug effects , Chlorambucil/chemistry , DNA/genetics , DNA/metabolism , Imidazoles/chemistry , Nylons/chemistry , Pyrroles/chemistry , Structure-Activity Relationship , Trinucleotide Repeats/drug effects
13.
Biochemistry (Mosc) ; 77(3): 270-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22803944

ABSTRACT

Trinucleotide repeat expansion provides a molecular basis for several devastating neurodegenerative diseases. In particular, expansion of a CAG run in the human HTT gene causes Huntington's disease. One of the main reasons for triplet repeat expansion in somatic cells is base excision repair (BER), involving damaged base excision and repair DNA synthesis that may be accompanied by expansion of the repaired strand due to formation of noncanonical DNA structures. We have analyzed the kinetics of excision of a ubiquitously found oxidized purine base, 8-oxoguanine (oxoG), by DNA glycosylase OGG1 from the substrates containing a CAG run flanked by AT-rich sequences. The values of k(2) rate constant for the removal of oxoG from triplets in the middle of the run were higher than for oxoG at the flanks of the run. The value of k(3) rate constant dropped starting from the third CAG-triplet in the run and remained stable until the 3'-terminal triplet, where it decreased even more. In nuclear extracts, the profile of oxoG removal rate along the run resembled the profile of k(2) constant, suggesting that the reaction rate in the extracts is limited by base excision. The fully reconstituted BER was efficient with all substrates unless oxoG was near the 3'-flank of the run, interfering with the initiation of the repair. DNA polymerase ß was able to perform a strand-displacement DNA synthesis, which may be important for CAG run expansion initiated by BER.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Guanine/analogs & derivatives , Trinucleotide Repeats/drug effects , Cell Line , DNA Glycosylases/genetics , Guanine/toxicity , Humans
14.
Bioorg Med Chem Lett ; 22(5): 2000-3, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22326165

ABSTRACT

Unusual expansion of trinucleotide repeats has been identified as a common mechanism of hereditary neurodegenerative diseases. Although the actual mechanism of repeat expansion remains uncertain, trinucleotide repeat instability may be related to the increased stability of an alternative DNA hairpin structure formed in the repeat sequences. Here we report that a synthetic ligand naphthyridine carbamate dimer (NCD) selectively bound to and stabilized an intra-stranded hairpin structure in CGG repeat sequences. The NCD-CGG hairpin complex was a stable structure that efficiently interfered with DNA replication by Taq DNA polymerase. Considering the sequence preference of NCD, the use of NCD would be valuable to investigate the genetic instabilities of CGG/CCG repeat sequences in human genomes.


Subject(s)
DNA/chemistry , Naphthyridines/chemistry , Naphthyridines/pharmacology , Nucleic Acid Conformation/drug effects , Trinucleotide Repeats/drug effects , Base Sequence , Carbamates/chemistry , Carbamates/pharmacology , DNA Replication/drug effects , Dimerization , Humans , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
15.
ACS Chem Biol ; 7(3): 496-505, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22252896

ABSTRACT

Recently, it was reported that expanded r(CAG) triplet repeats (r(CAG)(exp)) associated with untreatable neurological diseases cause pre-mRNA mis-splicing likely due to sequestration of muscleblind-like 1 (MBNL1) splicing factor. Bioactive small molecules that bind the 5'CAG/3'GAC motif found in r(CAG)(exp) hairpin structure were identified by using RNA binding studies and virtual screening/chemical similarity searching. Specifically, a benzylguanidine-containing small molecule was found to improve pre-mRNA alternative splicing of MBNL1-sensitive exons in cells expressing the toxic r(CAG)(exp). The compound was identified by first studying the binding of RNA 1 × 1 nucleotide internal loops to small molecules known to have affinity for nucleic acids. Those studies identified 4',6-diamidino-2-phenylindole (DAPI) as a specific binder to RNAs with the 5'CAG/3'GAC motif. DAPI was then used as a query molecule in a shape- and chemistry alignment-based virtual screen to identify compounds with improved properties, which identified 4-guanidinophenyl 4-guanidinobenzoate, a small molecule that improves pre-mRNA splicing defects associated with the r(CAG)(exp)-MBNL1 complex. This compound may facilitate the development of therapeutics to treat diseases caused by r(CAG)(exp) and could serve as a useful chemical tool to dissect the mechanisms of r(CAG)(exp) toxicity. The approach used in these studies, defining the small RNA motifs that bind small molecules with known affinity for nucleic acids and then using virtual screening to optimize them for bioactivity, may be generally applicable for designing small molecules that target other RNAs in the human genomic sequence.


Subject(s)
Benzoates/pharmacology , Guanidines/pharmacology , RNA Splicing/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Small Molecule Libraries/pharmacology , Transcription, Genetic/genetics , Trinucleotide Repeats/genetics , Benzoates/chemistry , Guanidines/chemistry , Humans , In Situ Hybridization, Fluorescence , Ligands , RNA Splicing/drug effects , RNA, Messenger/chemistry , RNA, Messenger/drug effects , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Transcription, Genetic/drug effects , Trinucleotide Repeats/drug effects , Tumor Cells, Cultured
16.
Chemistry ; 15(40): 10641-8, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19718722

ABSTRACT

A newly designed ligand, methylcarbamoylnaphthyridine dimer (MCND), was synthesized and characterized. Ligand binding to d(GAA)(10) was investigated by UV thermal denaturation, circular dichroism spectroscopy, surface plasmon resonance, and cold-spray-ionization time-of-flight mass spectrometry. The results indicated that MCND bound to the d(GAA)(n) repeat to form a stable hairpin structure with a major binding stoichiometry of 3:1. The most likely binding site was identified as the G-G mismatch in the AGA/AGA triad. The polymerase stop assay showed that MCND binding to the d(GAA)(n) repeat effectively interfered with the extension of the primer at the first two GAA sites on the template with both prokaryotic Taq DNA polymerase and human DNA polymerase alpha.


Subject(s)
DNA Replication , Models, Molecular , Trinucleotide Repeats/drug effects , Base Sequence , DNA Polymerase I/metabolism , Humans , Molecular Structure , Taq Polymerase/metabolism
17.
Bioorg Med Chem Lett ; 18(23): 6184-8, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18951785

ABSTRACT

Expansion of DNA repeat sequences is associated with many human genetic diseases. Bulged DNA structures have been implicated as intermediates in DNA slippage within the DNA repeat regions. Herein a bulge binding agent with novel wedge-shape topology of the aromatic moiety was designed and synthesized. The compound-bulge DNA interactions were characterized via UV melting experiments, circular dichroism and were quantitated by surface plasmon resonance with K(d) of 41.5 microM. This compound showed remarkable stimulation for DNA triplet repeat strand slippage synthesis in vitro.


Subject(s)
Amino Sugars/chemical synthesis , DNA Replication/drug effects , DNA/metabolism , Phenothiazines/chemical synthesis , Amino Sugars/chemistry , Amino Sugars/pharmacology , Binding Sites , DNA/chemistry , Humans , Molecular Structure , Phenothiazines/chemistry , Phenothiazines/pharmacology , Trinucleotide Repeats/drug effects
18.
PLoS One ; 3(2): e1595, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18270582

ABSTRACT

Non-coding CUG repeat expansions interfere with the activity of human Muscleblind-like (MBNL) proteins contributing to myotonic dystrophy 1 (DM1). To understand this toxic RNA gain-of-function mechanism we developed a Drosophila model expressing 60 pure and 480 interrupted CUG repeats in the context of a non-translatable RNA. These flies reproduced aspects of the DM1 pathology, most notably nuclear accumulation of CUG transcripts, muscle degeneration, splicing misregulation, and diminished Muscleblind function in vivo. Reduced Muscleblind activity was evident from the sensitivity of CUG-induced phenotypes to a decrease in muscleblind genetic dosage and rescue by MBNL1 expression, and further supported by the co-localization of Muscleblind and CUG repeat RNA in ribonuclear foci. Targeted expression of CUG repeats to the developing eye and brain mushroom bodies was toxic leading to rough eyes and semilethality, respectively. These phenotypes were utilized to identify genetic and chemical modifiers of the CUG-induced toxicity. 15 genetic modifiers of the rough eye phenotype were isolated. These genes identify putative cellular processes unknown to be altered by CUG repeat RNA, and they include mRNA export factor Aly, apoptosis inhibitor Thread, chromatin remodelling factor Nurf-38, and extracellular matrix structural component Viking. Ten chemical compounds suppressed the semilethal phenotype. These compounds significantly improved viability of CUG expressing flies and included non-steroidal anti-inflammatory agents (ketoprofen), muscarinic, cholinergic and histamine receptor inhibitors (orphenadrine), and drugs that can affect sodium and calcium metabolism such as clenbuterol and spironolactone. These findings provide new insights into the DM1 phenotype, and suggest novel candidates for DM1 treatments.


Subject(s)
DNA Repeat Expansion , Trinucleotide Repeats/drug effects , Trinucleotide Repeats/physiology , Animals , Brain , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Eye , Gene Dosage , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins
19.
Genes Cells ; 10(10): 953-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16164596

ABSTRACT

Fragile X syndrome is caused by expansion of a d(CGG) triplet repeat in the 5'-untranslated region of the first exon of the FMR1 gene resulting in silencing of the gene. The d(CGG) repeat has been reported to form hairpin and quadruplex structures in vitro, and formation of these higher structures could be responsible for its unstable expansion in the syndrome, although molecular mechanisms underlying the repeat expansion still remain elusive. We have previously proved that UP1, a proteolytic product of hnRNP A1, unfolds the intramolecular quadruplex structures of d(GGCAG)5 and d(TTAGGG)4 and abrogates the arrest of DNA synthesis at d(GGG)n sites. Here, we demonstrate that the d(CGG) repeat forms a peculiar DNA structure, which deviates from the canonical B-form structure. In addition, UP1 was demonstrated by CD spectrum analysis to unfold this characteristic higher structure of the d(CGG) repeat and to abrogate the arrest of DNA synthesis at the site. This ability of UP1 suggests that unfolding of unusual DNA structures of a triplet repeat is required for DNA synthesis processes.


Subject(s)
Nucleic Acid Conformation , Ribonucleoproteins/metabolism , Thymus Hormones/metabolism , Trinucleotide Repeats/drug effects , Circular Dichroism , DNA/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Electrophoretic Mobility Shift Assay , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Humans , Kinetics , Potassium Chloride/metabolism , Recombinant Proteins/metabolism , Ribonucleoproteins/pharmacology , Thymus Hormones/pharmacology , Trinucleotide Repeats/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...