Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(7): 5758-5782, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38511649

ABSTRACT

Eukaryotic translation initiation factor 2B (eIF2B) is a key component of the integrated stress response (ISR), which regulates protein synthesis and stress granule formation in response to cellular insult. Modulation of the ISR has been proposed as a therapeutic strategy for treatment of neurodegenerative diseases such as vanishing white matter (VWM) disease and amyotrophic lateral sclerosis (ALS) based on its ability to improve cellular homeostasis and prevent neuronal degeneration. Herein, we report the small-molecule discovery campaign that identified potent, selective, and CNS-penetrant eIF2B activators using both structure- and ligand-based drug design. These discovery efforts culminated in the identification of DNL343, which demonstrated a desirable preclinical drug profile, including a long half-life and high oral bioavailability across preclinical species. DNL343 was progressed into clinical studies and is currently undergoing evaluation in late-stage clinical trials for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Leukoencephalopathies , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Mutation , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Brain/metabolism , Leukoencephalopathies/metabolism
2.
Nucleic Acids Res ; 50(15): 8674-8689, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35904811

ABSTRACT

CGG repeat expansions in the FMR1 5'UTR cause the neurodegenerative disease Fragile X-associated tremor/ataxia syndrome (FXTAS). These repeats form stable RNA secondary structures that support aberrant translation in the absence of an AUG start codon (RAN translation), producing aggregate-prone peptides that accumulate within intranuclear neuronal inclusions and contribute to neurotoxicity. Here, we show that the most abundant RAN translation product, FMRpolyG, is markedly less toxic when generated from a construct with a non-repetitive alternating codon sequence in place of the CGG repeat. While exploring the mechanism of this differential toxicity, we observed a +1 translational frameshift within the CGG repeat from the arginine to glycine reading frame. Frameshifts occurred within the first few translated repeats and were triggered predominantly by RNA sequence and structural features. Short chimeric R/G peptides form aggregates distinct from those formed by either pure arginine or glycine, and these chimeras induce toxicity in cultured rodent neurons. Together, this work suggests that CGG repeats support translational frameshifting and that chimeric RAN translated peptides may contribute to CGG repeat-associated toxicity in FXTAS and related disorders.


Subject(s)
Fragile X Mental Retardation Protein , Neurodegenerative Diseases , Protein Aggregation, Pathological , Trinucleotide Repeats , Arginine/genetics , Ataxia , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome , Glycine/genetics , Humans , Neurodegenerative Diseases/genetics , Peptides/genetics , Peptides/metabolism , Recombinant Fusion Proteins/metabolism
3.
Acta Neuropathol Commun ; 8(1): 122, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753055

ABSTRACT

An intronic hexanucleotide repeat expansion in C9ORF72 causes familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This repeat is thought to elicit toxicity through RNA mediated protein sequestration and repeat-associated non-AUG (RAN) translation of dipeptide repeat proteins (DPRs). We generated a series of transgenic Drosophila models expressing GGGGCC (G4C2) repeats either inside of an artificial intron within a GFP reporter or within the 5' untranslated region (UTR) of GFP placed in different downstream reading frames. Expression of 484 intronic repeats elicited minimal alterations in eye morphology, viability, longevity, or larval crawling but did trigger RNA foci formation, consistent with prior reports. In contrast, insertion of repeats into the 5' UTR elicited differential toxicity that was dependent on the reading frame of GFP relative to the repeat. Greater toxicity correlated with a short and unstructured carboxyl terminus (C-terminus) in the glycine-arginine (GR) RAN protein reading frame. This change in C-terminal sequence triggered nuclear accumulation of all three RAN DPRs. A similar differential toxicity and dependence on the GR C-terminus was observed when repeats were expressed in rodent neurons. The presence of the native C-termini across all three reading frames was partly protective. Taken together, these findings suggest that C-terminal sequences outside of the repeat region may alter the behavior and toxicity of dipeptide repeat proteins derived from GGGGCC repeats.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , DNA Repeat Expansion , Frontotemporal Dementia/genetics , Protein Biosynthesis/genetics , Reading Frames/genetics , Animals , Dipeptides , Disease Models, Animal , Drosophila , Rats , Rats, Long-Evans
4.
Nat Neurosci ; 23(3): 386-397, 2020 03.
Article in English | MEDLINE | ID: mdl-32066985

ABSTRACT

Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.


Subject(s)
DNA Repeat Expansion/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Trinucleotide Repeats/genetics , Animals , Cell Line , Cell Survival/genetics , Female , Fragile X Mental Retardation Protein/biosynthesis , Induced Pluripotent Stem Cells , Male , Mice , Neurons/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics
5.
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
6.
EMBO Rep ; 20(9): e47498, 2019 09.
Article in English | MEDLINE | ID: mdl-31347257

ABSTRACT

A CGG trinucleotide repeat expansion in the 5' UTR of FMR1 causes the neurodegenerative disorder Fragile X-associated tremor/ataxia syndrome (FXTAS). This repeat supports a non-canonical mode of protein synthesis known as repeat-associated, non-AUG (RAN) translation. The mechanism underlying RAN translation at CGG repeats remains unclear. To identify modifiers of RAN translation and potential therapeutic targets, we performed a candidate-based screen of eukaryotic initiation factors and RNA helicases in cell-based assays and a Drosophila melanogaster model of FXTAS. We identified multiple modifiers of toxicity and RAN translation from an expanded CGG repeat in the context of the FMR1 5'UTR. These include the DEAD-box RNA helicase belle/DDX3X, the helicase accessory factors EIF4B/4H, and the start codon selectivity factors EIF1 and EIF5. Disrupting belle/DDX3X selectively inhibited FMR1 RAN translation in Drosophila in vivo and cultured human cells, and mitigated repeat-induced toxicity in Drosophila and primary rodent neurons. These findings implicate RNA secondary structure and start codon fidelity as critical elements mediating FMR1 RAN translation and identify potential targets for treating repeat-associated neurodegeneration.


Subject(s)
Ataxia/metabolism , DEAD-box RNA Helicases/metabolism , Drosophila Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Tremor/metabolism , Animals , Ataxia/genetics , Cells, Cultured , DEAD-box RNA Helicases/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Eukaryotic Initiation Factors/genetics , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Male , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Tremor/genetics
7.
Cell Rep ; 27(4): 1133-1150.e8, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31018129

ABSTRACT

The majority of individuals with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) exhibit neuronal cytoplasmic inclusions rich in the RNA binding protein TDP43. Even so, the relation between the RNA binding properties of TDP43 and neurodegeneration remains obscure. Here, we show that engineered mutations disrupting a salt bridge between the RNA recognition motifs of TDP43 interfere with RNA binding and eliminate the recognition of native TDP43 substrates. The same mutations dramatically destabilize TDP43, alter its subcellular localization, and abrogate TDP43-dependent neurodegeneration. Worms harboring homologous TDP-1 mutations phenocopy knockout strains, confirming the necessity of salt bridge residues for TDP43 function. Moreover, the accumulation of functional TDP43, but not RNA binding-deficient variants, disproportionately affects transcripts encoding ribosome and oxidative phosphorylation components. These studies demonstrate the significance of the salt bridge in sustaining TDP43 stability and RNA binding properties, factors that are crucial for neurodegeneration arising from TDP43 deposition in ALS and FTD.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mutation , Neurodegenerative Diseases/pathology , RNA/metabolism , Salts/chemistry , Animals , Caenorhabditis elegans , Female , Humans , Male , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Protein Aggregates , Protein Stability , RNA/chemistry , RNA, Mitochondrial/chemistry , RNA, Mitochondrial/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Rats
8.
Nat Commun ; 8(1): 2005, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222490

ABSTRACT

Repeat-associated non-AUG (RAN) translation allows for unconventional initiation at disease-causing repeat expansions. As RAN translation contributes to pathogenesis in multiple neurodegenerative disorders, determining its mechanistic underpinnings may inform therapeutic development. Here we analyze RAN translation at G4C2 repeat expansions that cause C9orf72-associated amyotrophic lateral sclerosis and frontotemporal dementia (C9RAN) and at CGG repeats that cause fragile X-associated tremor/ataxia syndrome. We find that C9RAN translation initiates through a cap- and eIF4A-dependent mechanism that utilizes a CUG start codon. C9RAN and CGG RAN are both selectively enhanced by integrated stress response (ISR) activation. ISR-enhanced RAN translation requires an eIF2α phosphorylation-dependent alteration in start codon fidelity. In parallel, both CGG and G4C2 repeats trigger phosphorylated-eIF2α-dependent stress granule formation and global translational suppression. These findings support a model whereby repeat expansions elicit cellular stress conditions that favor RAN translation of toxic proteins, creating a potential feed-forward loop that contributes to neurodegeneration.


Subject(s)
C9orf72 Protein/genetics , Neurodegenerative Diseases/genetics , Peptide Chain Initiation, Translational/genetics , Stress, Physiological/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Cell Extracts , Codon, Initiator/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4A/genetics , HEK293 Cells , HeLa Cells , Humans , Neurons , Phosphorylation/genetics , Primary Cell Culture , Rabbits , Rats , Reticulocytes
9.
PLoS One ; 11(10): e0165084, 2016.
Article in English | MEDLINE | ID: mdl-27776165

ABSTRACT

Hexanucleotide repeat expansions in C9orf72 are the most common inherited cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The expansions elicit toxicity in part through repeat-associated non-AUG (RAN) translation of the intronic (GGGGCC)n sequence into dipeptide repeat-containing proteins (DPRs). Little is known, however, about the structural characteristics and aggregation propensities of the dipeptide units comprising DPRs. To address this question, we synthesized dipeptide units corresponding to the three sense-strand RAN translation products, analyzed their structures by circular dichroism, electron microscopy and dye binding assays, and assessed their relative toxicity when applied to primary cortical neurons. Short, glycine-arginine (GR)3 dipeptides formed spherical aggregates and selectively reduced neuronal survival compared to glycine-alanine (GA)3 and glycine-proline (GP)3 dipeptides. Doubling peptide length had little effect on the structure of GR or GP peptides, but (GA)6 peptides formed ß-sheet rich aggregates that bound thioflavin T and Congo red yet lacked the typical fibrillar morphology of amyloids. Aging of (GA)6 dipeptides increased their ß-sheet content and enhanced their toxicity when applied to neurons. We also observed that the relative toxicity of each tested dipeptide was proportional to peptide internalization. Our results demonstrate that different C9orf72-related dipeptides exhibit distinct structural properties that correlate with their relative toxicity.


Subject(s)
DNA Repeat Expansion , Dipeptides/chemistry , Dipeptides/toxicity , Neurons/cytology , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein , Cell Survival/drug effects , Cells, Cultured , Circular Dichroism , Frontotemporal Dementia/genetics , Genetic Predisposition to Disease , Humans , Models, Molecular , Mutation , Neurons/drug effects , Protein Structure, Secondary , Rats
10.
Cell Rep ; 12(7): 1169-83, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26257172

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous distal symmetric polyneuropathy. Whole-exome sequencing (WES) of 40 individuals from 37 unrelated families with CMT-like peripheral neuropathy refractory to molecular diagnosis identified apparent causal mutations in ∼ 45% (17/37) of families. Three candidate disease genes are proposed, supported by a combination of genetic and in vivo studies. Aggregate analysis of mutation data revealed a significantly increased number of rare variants across 58 neuropathy-associated genes in subjects versus controls, confirmed in a second ethnically discrete neuropathy cohort, suggesting that mutation burden potentially contributes to phenotypic variability. Neuropathy genes shown to have highly penetrant Mendelizing variants (HPMVs) and implicated by burden in families were shown to interact genetically in a zebrafish assay exacerbating the phenotype established by the suppression of single genes. Our findings suggest that the combinatorial effect of rare variants contributes to disease burden and variable expressivity.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Exome , Genetic Load , Peripheral Nervous System Diseases/genetics , Phenotype , Animals , Female , Genetic Variation , HSP40 Heat-Shock Proteins/genetics , Humans , Male , Mutation , Myelin P2 Protein/genetics , Pedigree , Penetrance , Serine C-Palmitoyltransferase/genetics , Suppression, Genetic , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...