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










Publication year range
1.
Eur J Neurosci ; 53(8): 2450-2468, 2021 04.
Article in English | MEDLINE | ID: mdl-33759265

ABSTRACT

Disruptive or excessive repetitive motor patterns (stereotypies) are cardinal symptoms in numerous neuropsychiatric disorders. Stereotypies are also evoked by psychomotor stimulants such as amphetamine. The acquisition of motor sequences is paralleled by changes in activity patterns in the striatum, and stereotypies have been linked to abnormal plasticity in these reinforcement-related circuits. Here, we designed experiments in mice to identify transcriptomic changes that underlie striatal plasticity occurring alongside the development of drug-induced stereotypic behavior. We identified three schedules of amphetamine treatment inducing different degrees of stereotypy and used bulk RNAseq to compare striatal gene expression changes among groups of mice treated with the different drug-dose schedules and vehicle-treated, cage-mate controls. Mice were identified as naïve, sensitized, or tolerant to drug-induced stereotypy. All drug-treated groups exhibited expression changes in genes that encode members of the extracellular signal-regulated kinase (ERK) cascades known to regulate psychomotor stimulant responses. In the sensitized group with the most prolonged stereotypy, we found dysregulation of 20 genes that were not changed in other groups. Gene set enrichment analysis indicated highly significant overlap with genes regulated by neuregulin 1 (Nrg1). Nrg1 is known to be a schizophrenia and autism susceptibility gene that encodes a ligand for Erb-B receptors, which are involved in neuronal migration, myelination, and cell survival, including that of dopamine-containing neurons. Stimulant abuse is a risk factor for schizophrenia onset, and these two disorders share behavioral stereotypy phenotypes. Our results raise the possibility that drug-induced sensitization of the Nrg1 signaling pathway might underlie these links.


Subject(s)
Pharmaceutical Preparations , Transcriptome , Amphetamine , Animals , Corpus Striatum , Mice , Stereotyped Behavior
2.
Proc Natl Acad Sci U S A ; 116(49): 24840-24851, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31744868

ABSTRACT

Huntington's disease (HD) is a chronic neurodegenerative disorder characterized by a late clinical onset despite ubiquitous expression of the mutant Huntingtin gene (HTT) from birth. Transcriptional dysregulation is a pivotal feature of HD. Yet, the genes that are altered in the prodromal period and their regulators, which present opportunities for therapeutic intervention, remain to be elucidated. Using transcriptional and chromatin profiling, we found aberrant transcription and changes in histone H3K27acetylation in the striatum of R6/1 mice during the presymptomatic disease stages. Integrating these data, we identified the Elk-1 transcription factor as a candidate regulator of prodromal changes in HD. Exogenous expression of Elk-1 exerted beneficial effects in a primary striatal cell culture model of HD, and adeno-associated virus-mediated Elk-1 overexpression alleviated transcriptional dysregulation in R6/1 mice. Collectively, our work demonstrates that aberrant gene expression precedes overt disease onset in HD, identifies the Elk-1 transcription factor as a key regulator linked to early epigenetic and transcriptional changes in HD, and presents evidence for Elk-1 as a target for alleviating molecular pathology in HD.


Subject(s)
Epigenomics , Huntington Disease/genetics , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism , Animals , Corpus Striatum/metabolism , Dependovirus , Disease Models, Animal , Histones/metabolism , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Mice , Mice, Transgenic , Neurons/metabolism , Nuclear Proteins/metabolism
3.
J Clin Invest ; 128(8): 3630-3641, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29809168

ABSTRACT

Skeletal muscle has emerged as a critical, disease-relevant target tissue in spinal and bulbar muscular atrophy, a degenerative disorder of the neuromuscular system caused by a CAG/polyglutamine (polyQ) expansion in the androgen receptor (AR) gene. Here, we used RNA-sequencing (RNA-Seq) to identify pathways that are disrupted in diseased muscle using AR113Q knockin mice. This analysis unexpectedly identified substantially diminished expression of numerous ubiquitin/proteasome pathway genes in AR113Q muscle, encoding approximately 30% of proteasome subunits and 20% of E2 ubiquitin conjugases. These changes were age, hormone, and glutamine length dependent and arose due to a toxic gain of function conferred by the mutation. Moreover, altered gene expression was associated with decreased levels of the proteasome transcription factor NRF1 and its activator DDI2 and resulted in diminished proteasome activity. Ubiquitinated ADRM1 was detected in AR113Q muscle, indicating the occurrence of stalled proteasomes in mutant mice. Finally, diminished expression of Drosophila orthologues of NRF1 or ADRM1 promoted the accumulation of polyQ AR protein and increased toxicity. Collectively, these data indicate that AR113Q muscle develops progressive proteasome dysfunction that leads to the impairment of quality control and the accumulation of polyQ AR protein, key features that contribute to the age-dependent onset and progression of this disorder.


Subject(s)
Aging/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Peptides/metabolism , Receptors, Androgen/metabolism , Trinucleotide Repeat Expansion , Aging/genetics , Aging/pathology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peptides/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Receptors, Androgen/genetics
4.
Sci Rep ; 7(1): 12556, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28970536

ABSTRACT

The heat shock response (HSR) is a mechanism to cope with proteotoxic stress by inducing the expression of molecular chaperones and other heat shock response genes. The HSR is evolutionarily well conserved and has been widely studied in bacteria, cell lines and lower eukaryotic model organisms. However, mechanistic insights into the HSR in higher eukaryotes, in particular in mammals, are limited. We have developed an in vivo heat shock protocol to analyze the HSR in mice and dissected heat shock factor 1 (HSF1)-dependent and -independent pathways. Whilst the induction of proteostasis-related genes was dependent on HSF1, the regulation of circadian function related genes, indicating that the circadian clock oscillators have been reset, was independent of its presence. Furthermore, we demonstrate that the in vivo HSR is impaired in mouse models of Huntington's disease but we were unable to corroborate the general repression of transcription that follows a heat shock in lower eukaryotes.


Subject(s)
Heat Shock Transcription Factors/genetics , Heat-Shock Response/genetics , Huntington Disease/genetics , Animals , Circadian Clocks/genetics , DNA-Binding Proteins , Disease Models, Animal , Humans , Huntington Disease/pathology , Mice
5.
Cell Rep ; 19(7): 1365-1377, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514657

ABSTRACT

Brain microvascular endothelial cells (BMECs) are an essential component of the blood-brain barrier (BBB) that shields the brain against toxins and immune cells. While BBB dysfunction exists in neurological disorders, including Huntington's disease (HD), it is not known if BMECs themselves are functionally compromised to promote BBB dysfunction. Further, the underlying mechanisms of BBB dysfunction remain elusive given limitations with mouse models and post-mortem tissue to identify primary deficits. We undertook a transcriptome and functional analysis of human induced pluripotent stem cell (iPSC)-derived BMECs (iBMEC) from HD patients or unaffected controls. We demonstrate that HD iBMECs have intrinsic abnormalities in angiogenesis and barrier properties, as well as in signaling pathways governing these processes. Thus, our findings provide an iPSC-derived BBB model for a neurodegenerative disease and demonstrate autonomous neurovascular deficits that may underlie HD pathology with implications for therapeutics and drug delivery.


Subject(s)
Blood-Brain Barrier/pathology , Endothelial Cells/pathology , Huntington Disease/pathology , Induced Pluripotent Stem Cells/pathology , Microvessels/pathology , Neovascularization, Physiologic , Wnt Signaling Pathway , Gene Regulatory Networks , Humans , Huntington Disease/genetics , Induced Pluripotent Stem Cells/metabolism , Transcriptome/genetics , Transcytosis , beta Catenin/metabolism
6.
RNA Biol ; 10(11): 1647-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24256709

ABSTRACT

Huntington's disease (HD) is an adult-onset neurodegenerative disorder caused by a mutated CAG repeat in the huntingtin gene that is translated into an expanded polyglutamine tract. The clinical manifestation of HD is a progressive physical, cognitive, and psychiatric deterioration that is eventually fatal. The mutant huntingtin protein is processed into several smaller fragments, which have been implicated as critical factors in HD pathogenesis. The search for proteases responsible for their production has led to the identification of several cleavage sites on the huntingtin protein. However, the origin of the small N-terminal fragments that are found in HD postmortem brains has remained elusive. Recent mapping of huntingtin fragments in a mouse model demonstrated that the smallest N-terminal fragment is an exon 1 protein. This discovery spurred our hypothesis that mis-splicing as opposed to proteolysis could be generating the smallest huntingtin fragment. We demonstrated that mis-splicing of mutant huntingtin intron 1 does indeed occur and results in a short polyadenylated mRNA, which is translated into an exon 1 protein. The exon 1 protein fragment is highly pathogenic. Transgenic mouse models containing just human huntingtin exon 1 develop a rapid onset of HD-like symptoms. Our finding that a small, mis-spliced HTT transcript and corresponding exon 1 protein are produced in the context of an expanded CAG repeat has unraveled a new molecular mechanism in HD pathogenesis. Here we present detailed models of how mis-splicing could be facilitated, what challenges remain in this model, and implications for therapeutic studies.


Subject(s)
Huntington Disease/genetics , Mutant Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA Splicing , Animals , Humans
7.
Proc Natl Acad Sci U S A ; 110(32): E3027-36, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23872847

ABSTRACT

Transcriptional dysregulation is an early feature of Huntington disease (HD). We observed gene-specific changes in histone H3 lysine 4 trimethylation (H3K4me3) at transcriptionally repressed promoters in R6/2 mouse and human HD brain. Genome-wide analysis showed a chromatin signature for this mark. Reducing the levels of the H3K4 demethylase SMCX/Jarid1c in primary neurons reversed down-regulation of key neuronal genes caused by mutant Huntingtin expression. Finally, reduction of SMCX/Jarid1c in primary neurons from BACHD mice or the single Jarid1 in a Drosophila HD model was protective. Therefore, targeting this epigenetic signature may be an effective strategy to ameliorate the consequences of HD.


Subject(s)
Brain/metabolism , Histones/metabolism , Huntington Disease/metabolism , Lysine/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Expression Profiling , Histone Demethylases , Humans , Huntingtin Protein , Huntington Disease/genetics , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 110(6): 2366-70, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23341618

ABSTRACT

Huntington disease (HD) is a devastating, late-onset, inherited neurodegenerative disorder that manifests with personality changes, movement disorders, and cognitive decline. It is caused by a CAG repeat expansion in exon 1 of the HTT gene that translates to a polyglutamine tract in the huntingtin protein (HTT). The formation of HTT fragments has been implicated as an essential step in the molecular pathogenesis of HD and several proteases that cleave HTT have been identified. However, the importance of smaller N-terminal fragments has been highlighted by their presence in HD postmortem brains and by the fact that nuclear inclusions are only detected by antibodies to the N terminus of HTT. Despite an intense research effort, the precise length of these fragments and the mechanism by which they are generated remains unknown. Here we show that CAG repeat length-dependent aberrant splicing of exon 1 HTT results in a short polyadenylated mRNA that is translated into an exon 1 HTT protein. Given that mutant exon 1 HTT proteins have consistently been shown to be highly pathogenic in HD mouse models, the aberrant splicing of HTT mRNA provides a mechanistic basis for the molecular pathogenesis of HD. RNA-targeted therapeutic strategies designed to lower the levels of HTT are under development. Many of these approaches would not prevent the production of exon 1 HTT and should be reviewed in light of our findings.


Subject(s)
Huntington Disease/genetics , Mutant Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , RNA Splicing , Animals , Base Sequence , Brain/metabolism , Disease Models, Animal , Exons , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Molecular Sequence Data , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Trinucleotide Repeats
9.
Methods Mol Biol ; 714: 423-44, 2011.
Article in English | MEDLINE | ID: mdl-21431756

ABSTRACT

RNA affinity tags (aptamers) have emerged as useful tools for the isolation of RNAs and ribonucleoprotein complexes from cell extracts. The streptavidin binding RNA aptamer binds with high affinity and is quickly and cleanly eluted with biotin under mild conditions that retain intact complexes. We describe the use of the streptavidin binding aptamer as a tool for purification and discuss strategies towards the design and production of tagged RNAs with a focus on structured target RNAs. The aptamer site can be further exploited as a unique region for the hybridization of oligonucleotide probes and localization by fluorescent in situ hybridization (FISH). The aptamer insertion will allow the localization of a population of RNA species (such as mutants) to be viewed specifically, while in the presence of the wild type RNA. We describe the production of labeled oligonucleotide probes and the preparation of yeast cells for the localization of RNAs by FISH.


Subject(s)
Aptamers, Nucleotide/metabolism , RNA/isolation & purification , RNA/metabolism , Ribonucleoproteins/isolation & purification , Ribonucleoproteins/metabolism , Base Sequence , In Situ Hybridization, Fluorescence , Macromolecular Substances/chemistry , Macromolecular Substances/isolation & purification , Macromolecular Substances/metabolism , Molecular Sequence Data , Oligonucleotide Probes/genetics , Permeability , Polymerase Chain Reaction , Protein Transport , RNA/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Streptavidin/metabolism
10.
Genes Dev ; 22(16): 2204-14, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18708579

ABSTRACT

The 274 tRNA genes in Saccharomyces cerevisiae are scattered throughout the linear maps of the 16 chromosomes, but the genes are clustered at the nucleolus when compacted in the nucleus. This clustering is dependent on intact nucleolar organization and contributes to tRNA gene-mediated (tgm) silencing of RNA polymerase II transcription near tRNA genes. After examination of the localization mechanism, we find that the chromosome-condensing complex, condensin, is involved in the clustering of tRNA genes. Conditionally defective mutations in all five subunits of condensin, which we confirm is bound to active tRNA genes in the yeast genome, lead to loss of both pol II transcriptional silencing near tRNA genes and nucleolar clustering of the genes. Furthermore, we show that condensin physically associates with a subcomplex of RNA polymerase III transcription factors on the tRNA genes. Clustering of tRNA genes by condensin appears to be a separate mechanism from their nucleolar localization, as microtubule disruption releases tRNA gene clusters from the nucleolus, but does not disperse the clusters. These observations suggest a widespread role for condensin in gene organization and packaging of the interphase yeast nucleus.


Subject(s)
Adenosine Triphosphatases/physiology , Cell Nucleolus/physiology , DNA-Binding Proteins/physiology , Multiprotein Complexes/physiology , RNA, Transfer/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Cell Cycle/drug effects , Cell Cycle/physiology , Chromatin Immunoprecipitation , Gene Silencing , Genome, Fungal , In Situ Hybridization , Interphase/physiology , Microtubules/metabolism , Mutation/genetics , Nocodazole/pharmacology , Polymerase Chain Reaction , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , RNA, Fungal/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...