Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(7): 114375, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38935506

ABSTRACT

GGGGCC (G4C2) repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). How this genetic mutation leads to neurodegeneration remains largely unknown. Using CRISPR-Cas9 technology, we deleted EXOC2, which encodes an essential exocyst subunit, in induced pluripotent stem cells (iPSCs) derived from C9ORF72-ALS/FTD patients. These cells are viable owing to the presence of truncated EXOC2, suggesting that exocyst function is partially maintained. Several disease-relevant cellular phenotypes in C9ORF72 iPSC-derived motor neurons are rescued due to, surprisingly, the decreased levels of dipeptide repeat (DPR) proteins and expanded G4C2 repeats-containing RNA. The treatment of fully differentiated C9ORF72 neurons with EXOC2 antisense oligonucleotides also decreases expanded G4C2 repeats-containing RNA and partially rescued disease phenotypes. These results indicate that EXOC2 directly or indirectly regulates the level of G4C2 repeats-containing RNA, making it a potential therapeutic target in C9ORF72-ALS/FTD.

2.
Elife ; 122023 09 07.
Article in English | MEDLINE | ID: mdl-37675986

ABSTRACT

A hexanucleotide repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). A hallmark of ALS/FTD pathology is the presence of dipeptide repeat (DPR) proteins, produced from both sense GGGGCC (poly-GA, poly-GP, poly-GR) and antisense CCCCGG (poly-PR, poly-PG, poly-PA) transcripts. Translation of sense DPRs, such as poly-GA and poly-GR, depends on non-canonical (non-AUG) initiation codons. Here, we provide evidence for canonical AUG-dependent translation of two antisense DPRs, poly-PR and poly-PG. A single AUG is required for synthesis of poly-PR, one of the most toxic DPRs. Unexpectedly, we found redundancy between three AUG codons necessary for poly-PG translation. Further, the eukaryotic translation initiation factor 2D (EIF2D), which was previously implicated in sense DPR synthesis, is not required for AUG-dependent poly-PR or poly-PG translation, suggesting that distinct translation initiation factors control DPR synthesis from sense and antisense transcripts. Our findings on DPR synthesis from the C9ORF72 locus may be broadly applicable to many other nucleotide repeat expansion disorders.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , Frontotemporal Dementia , Pick Disease of the Brain , Humans , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Codon, Initiator/genetics , Dipeptides/genetics , Dipeptides/metabolism , Frontotemporal Dementia/pathology , Proteins/genetics
3.
Nat Commun ; 13(1): 55, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013139

ABSTRACT

Although the synaptic alterations associated with the stress-related mood disorder major depression has been well-documented, the underlying transcriptional mechanisms remain poorly understood. Here, we perform complementary bulk nuclei- and single-nucleus transcriptome profiling and map locus-specific chromatin interactions in mouse neocortex to identify the cell type-specific transcriptional changes associated with stress-induced behavioral maladaptation. We find that cortical excitatory neurons, layer 2/3 neurons in particular, are vulnerable to chronic stress and acquire signatures of gene transcription and chromatin structure associated with reduced neuronal activity and expression of Yin Yang 1 (YY1). Selective ablation of YY1 in cortical excitatory neurons enhances stress sensitivity in both male and female mice and alters the expression of stress-associated genes following an abbreviated stress exposure. These findings demonstrate how chronic stress impacts transcription in cortical excitatory neurons and identify YY1 as a regulator of stress-induced maladaptive behavior in mice.


Subject(s)
Neurons/metabolism , Prefrontal Cortex/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism , Animals , Behavior, Animal , Chromatin/metabolism , Epigenomics , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stress, Physiological
4.
Genome Res ; 28(7): 933-942, 2018 07.
Article in English | MEDLINE | ID: mdl-29848492

ABSTRACT

Genetic variants associated with autism spectrum disorders (ASDs) are enriched in genes encoding synaptic proteins and chromatin regulators. Although the role of synaptic proteins in ASDs is widely studied, the mechanism by which chromatin regulators contribute to ASD risk remains poorly understood. Upon profiling and analyzing the transcriptional and epigenomic features of genes expressed in the cortex, we uncovered a unique set of long genes that contain broad enhancer-like chromatin domains (BELDs) spanning across their entire gene bodies. Analyses of these BELD genes show that they are highly transcribed with frequent RNA polymerase II (Pol II) initiation and low Pol II pausing, and they exhibit frequent chromatin-chromatin interactions within their gene bodies. These BELD features are conserved from rodents to humans, are enriched in genes involved in synaptic function, and appear post-natally concomitant with synapse development. Importantly, we find that BELD genes are highly implicated in neurodevelopmental disorders, particularly ASDs, and that their expression is preferentially down-regulated in individuals with idiopathic autism. Finally, we find that the transcription of BELD genes is particularly sensitive to alternations in ASD-associated chromatin regulators. These findings suggest that the epigenomic regulation of BELD genes is important for post-natal cortical development and lend support to a model by which mutations in chromatin regulators causally contribute to ASDs by preferentially impairing BELD gene transcription.


Subject(s)
Autism Spectrum Disorder/genetics , Chromatin/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Autistic Disorder/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Neurogenesis/genetics , RNA Polymerase II/genetics , Transcription, Genetic/genetics
5.
Mol Cell ; 68(5): 1006-1015.e7, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29220646

ABSTRACT

Massively parallel single-cell RNA sequencing can precisely resolve cellular diversity in a high-throughput manner at low cost, but unbiased isolation of intact single cells from complex tissues such as adult mammalian brains is challenging. Here, we integrate sucrose-gradient-assisted purification of nuclei with droplet microfluidics to develop a highly scalable single-nucleus RNA-seq approach (sNucDrop-seq), which is free of enzymatic dissociation and nucleus sorting. By profiling ∼18,000 nuclei isolated from cortical tissues of adult mice, we demonstrate that sNucDrop-seq not only accurately reveals neuronal and non-neuronal subtype composition with high sensitivity but also enables in-depth analysis of transient transcriptional states driven by neuronal activity, at single-cell resolution, in vivo.


Subject(s)
Cell Nucleus/metabolism , Cerebral Cortex/metabolism , High-Throughput Nucleotide Sequencing , Neurons/metabolism , RNA/genetics , Seizures/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcription, Genetic , Animals , Cell Nucleus/pathology , Centrifugation, Density Gradient , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , Human Embryonic Stem Cells/metabolism , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Microfluidic Analytical Techniques , NIH 3T3 Cells , Neural Inhibition , Neurons/pathology , Pentylenetetrazole , RNA/metabolism , Seizures/metabolism , Seizures/pathology , Seizures/physiopathology , Synaptic Transmission , Transfection
6.
Nat Commun ; 8: 15315, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497787

ABSTRACT

Efforts to manipulate locus-specific histone acetylation to assess their causal role in gene expression and cellular and behavioural phenotypes have been impeded by a lack of experimental tools. The Cas9 nuclease has been adapted to target epigenomic modifications, but a detailed description of the parameters of such synthetic epigenome remodellers is still lacking. Here we describe a Cas9-based histone deacetylase (HDAC) and the design principles required to achieve locus-specific histone deacetylation. We assess its range of activity and specificity, and analyse target gene expression in two different cell types to investigate cellular context-dependent effects. Our findings demonstrate that the chromatin environment is an important element to consider when utilizing this synthetic HDAC.


Subject(s)
CRISPR-Cas Systems , Chromatin/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Acetylation , Animals , Cell Line, Tumor , Chromatin/genetics , Epigenomics/methods , Gene Expression Profiling , Gene Expression Regulation , Genetic Loci , HEK293 Cells , Histone Deacetylases/genetics , Humans , Mice , Promoter Regions, Genetic/genetics
7.
J Clin Invest ; 127(5): 1889-1904, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28394263

ABSTRACT

Mutations in the X-linked gene encoding methyl-CpG-binding protein 2 (MeCP2) cause Rett syndrome (RTT), a neurological disorder affecting cognitive development, respiration, and motor function. Genetic restoration of MeCP2 expression reverses RTT-like phenotypes in mice, highlighting the need to search for therapeutic approaches. Here, we have developed knockin mice recapitulating the most common RTT-associated missense mutation, MeCP2 T158M. We found that the T158M mutation impaired MECP2 binding to methylated DNA and destabilized MeCP2 protein in an age-dependent manner, leading to the development of RTT-like phenotypes in these mice. Genetic elevation of MeCP2 T158M expression ameliorated multiple RTT-like features, including motor dysfunction and breathing irregularities, in both male and female mice. These improvements were accompanied by increased binding of MeCP2 T158M to DNA. Further, we found that the ubiquitin/proteasome pathway was responsible for MeCP2 T158M degradation and that proteasome inhibition increased MeCP2 T158M levels. Together, these findings demonstrate that increasing MeCP2 T158M protein expression is sufficient to mitigate RTT-like phenotypes and support the targeting of MeCP2 T158M expression or stability as an alternative therapeutic approach.


Subject(s)
Gene Expression Regulation , Methyl-CpG-Binding Protein 2 , Mutation, Missense , Proteolysis , Rett Syndrome , Amino Acid Substitution , Animals , Humans , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rett Syndrome/pathology , Ubiquitin/genetics , Ubiquitin/metabolism
8.
Neurobiol Dis ; 88: 118-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792401

ABSTRACT

The development of therapeutics for neurological disorders is constrained by limited access to the central nervous system (CNS). ATP-binding cassette (ABC) transporters, particularly P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), are expressed on the luminal surface of capillaries in the CNS and transport drugs out of the endothelium back into the blood against the concentration gradient. Survival motor neuron (SMN) protein, which is deficient in spinal muscular atrophy (SMA), is a target of the ubiquitin proteasome system. Inhibiting the proteasome in a rodent model of SMA with bortezomib increases SMN protein levels in peripheral tissues but not the CNS, because bortezomib has poor CNS penetrance. We sought to determine if we could inhibit SMN degradation in the CNS of SMA mice with a combination of bortezomib and the ABC transporter inhibitor tariquidar. In cultured cells we show that bortezomib is a substrate of P-gp. Mass spectrometry analysis demonstrated that intraperitoneal co-administration of tariquidar increased the CNS penetrance of bortezomib, and reduced proteasome activity in the brain and spinal cord. This correlated with increased SMN protein levels and improved survival and motor function of SMA mice. These findings show that CNS penetrance of treatment for this neurological disorder can be improved by inhibiting drug efflux at the blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/metabolism , Bortezomib/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Age Factors , Animals , Animals, Newborn , Antineoplastic Agents/pharmacology , Central Nervous System/cytology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Transgenic , Motor Neurons/drug effects , Proteasome Endopeptidase Complex , Quinolines/pharmacology , Quinolines/therapeutic use , Time Factors , Transfection
10.
Mol Biol Cell ; 24(12): 1863-71, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23615451

ABSTRACT

Spinal muscular atrophy is an inherited motor neuron disease that results from a deficiency of the survival of motor neuron (SMN) protein. SMN is ubiquitinated and degraded through the ubiquitin proteasome system (UPS). We have previously shown that proteasome inhibition increases SMN protein levels, improves motor function, and reduces spinal cord, muscle, and neuromuscular junction pathology of spinal muscular atrophy (SMA) mice. Specific targets in the UPS may be more efficacious and less toxic. In this study, we show that the E3 ubiquitin ligase, mind bomb 1 (Mib1), interacts with and ubiquitinates SMN and facilitates its degradation. Knocking down Mib1 levels increases SMN protein levels in cultured cells. Also, knocking down the Mib1 orthologue improves neuromuscular function in Caenorhabditis elegans deficient in SMN. These findings demonstrate that Mib1 ubiquitinates and catalyzes the degradation of SMN, and thus represents a novel therapeutic target for SMA.


Subject(s)
Survival of Motor Neuron 1 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , HEK293 Cells , Humans , Hybrid Cells , Mice , Neuroblastoma/pathology , Pharyngeal Muscles/metabolism , Pharyngeal Muscles/physiopathology , Protein Binding , Proteolysis , RNA Interference , Spinal Cord/cytology , Survival of Motor Neuron 1 Protein/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
11.
PLoS One ; 7(1): e30437, 2012.
Article in English | MEDLINE | ID: mdl-22276200

ABSTRACT

P-element vectors are commonly used to make transgenic Drosophila and generally insert in the genome in a nonselective manner. However, when specific fragments of regulatory DNA from a few Drosophila genes are incorporated into P-transposons, they cause the vectors to be inserted near the gene from which the DNA fragment was derived. This is called P-element homing. We mapped the minimal DNA fragment that could mediate homing to the engrailed/invected region of the genome. A 1.6 kb fragment of engrailed regulatory DNA that contains two Polycomb-group response elements (PREs) was sufficient for homing. We made flies that contain a 1.5 kb deletion of engrailed DNA (en(Δ1.5)) in situ, including the PREs and the majority of the fragment that mediates homing. Remarkably, homing still occurs onto the en(Δ1. 5) chromosome. In addition to homing to en, P[en] inserts near Polycomb group target genes at an increased frequency compared to P[EPgy2], a vector used to generate 18,214 insertions for the Drosophila gene disruption project. We suggest that homing is mediated by interactions between multiple proteins bound to the homing fragment and proteins bound to multiple areas of the engrailed/invected chromatin domain. Chromatin structure may also play a role in homing.


Subject(s)
Drosophila Proteins/genetics , Genetic Vectors/genetics , Repressor Proteins/genetics , Response Elements/genetics , Animals , Chromosomes/genetics , Drosophila melanogaster , Polycomb-Group Proteins
12.
Hum Mol Genet ; 20(18): 3667-77, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21693563

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by reduced levels of the survival motor neuron (SMN) protein. Here we show that the proteasome inhibitor, bortezomib, increases SMN in cultured cells and in peripheral tissues of SMA model mice. Bortezomib-treated animals had improved motor function, which was associated with reduced spinal cord and muscle pathology and improved neuromuscular junction size, but no change in survival. Combining bortezomib with the histone deacetylase inhibitor trichostatin A (TSA) resulted in a synergistic increase in SMN protein levels in mouse tissue and extended survival of SMA mice more than TSA alone. Our results demonstrate that a combined regimen of drugs that decrease SMN protein degradation and increase SMN gene transcription synergistically increases SMN levels and improves the lifespan of SMA model mice. Moreover, this study indicates that while increasing SMN levels in the central nervous system may help extend survival, peripheral tissues can also be targeted to improve the SMA disease phenotype.


Subject(s)
Down-Regulation , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Up-Regulation , Animals , Cells, Cultured , Child, Preschool , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/pathology , Phenotype , Proteolysis , Survival
13.
Mol Cell Biol ; 29(5): 1107-15, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19103745

ABSTRACT

Spinal muscular atrophy (SMA) is caused by mutations of the survival of motor neuron (SMN1) gene and deficiency of full-length SMN protein (FL-SMN). All SMA patients retain one or more copies of the SMN2 gene, but the principal protein product of SMN2 lacks exon 7 (SMNDelta7) and is unable to compensate for a deficiency of FL-SMN. SMN is known to oligomerize and form a multimeric protein complex; however, the mechanisms regulating stability and degradation of FL-SMN and SMNDelta7 proteins have been largely unexplored. Using pulse-chase analysis, we characterized SMN protein turnover and confirmed that SMN was ubiquitinated and degraded by the ubiquitin proteasome system (UPS). The SMNDelta7 protein had a twofold shorter half-life than FL-SMN in cells despite similar intrinsic rates of turnover by the UPS in a cell-free assay. Mutations that inhibited SMN oligomerization and complex formation reduced the FL-SMN half-life. Furthermore, recruitment of SMN into large macromolecular complexes as well as increased association with several Gemin proteins was regulated in part by protein kinase A. Together, our data indicate that SMN protein stability is modulated by complex formation. Promotion of the SMN complex formation may be an important novel therapeutic strategy for SMA.


Subject(s)
Mutation , Survival of Motor Neuron 1 Protein/metabolism , Dimerization , Half-Life , Humans , Multiprotein Complexes , Proteasome Endopeptidase Complex , Protein Stability , Survival of Motor Neuron 1 Protein/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...