Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neuron ; 112(13): 2112-2129.e4, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38692279

ABSTRACT

Genome-wide association studies (GWASs) have uncovered over 75 genomic loci associated with risk for late-onset Alzheimer's disease (LOAD), but identification of the underlying causal genes remains challenging. Studies of induced pluripotent stem cell (iPSC)-derived neurons from LOAD patients have demonstrated the existence of neuronal cell-intrinsic functional defects. Here, we searched for genetic contributions to neuronal dysfunction in LOAD using an integrative systems approach that incorporated multi-evidence-based gene mapping and network-analysis-based prioritization. A systematic perturbation screening of candidate risk genes in Caenorhabditis elegans (C. elegans) revealed that neuronal knockdown of the LOAD risk gene orthologs vha-10 (ATP6V1G2), cmd-1 (CALM3), amph-1 (BIN1), ephx-1 (NGEF), and pho-5 (ACP2) alters short-/intermediate-term memory function, the cognitive domain affected earliest during LOAD progression. These results highlight the impact of LOAD risk genes on evolutionarily conserved memory function, as mediated through neuronal endosomal dysfunction, and identify new targets for further mechanistic interrogation.


Subject(s)
Alzheimer Disease , Caenorhabditis elegans , Genome-Wide Association Study , Alzheimer Disease/genetics , Caenorhabditis elegans/genetics , Animals , Humans , Systems Biology/methods , Memory/physiology , Induced Pluripotent Stem Cells , Neurons/metabolism , Genetic Predisposition to Disease/genetics , Caenorhabditis elegans Proteins/genetics
2.
bioRxiv ; 2024 May 02.
Article in English | MEDLINE | ID: mdl-38260399

ABSTRACT

RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.

3.
Biochem Soc Trans ; 51(1): 161-171, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36744642

ABSTRACT

How organisms learn and encode memory is an outstanding question in neuroscience research. Specifically, how memories are acquired and consolidated at the level of molecular and gene pathways remains unclear. In addition, memory is disrupted in a wide variety of neurological disorders; therefore, discovering molecular regulators of memory may reveal therapeutic targets for these disorders. C. elegans are an excellent model to uncover molecular and genetic regulators of memory. Indeed, the nematode's invariant neuronal lineage, fully mapped genome, and conserved associative behaviors have allowed the development of a breadth of genetic and genomic tools to examine learning and memory. In this mini-review, we discuss novel and exciting genetic and genomic techniques used to examine molecular and genetic underpinnings of memory from the level of the whole-worm to tissue-specific and cell-type specific approaches with high spatiotemporal resolution.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Learning/physiology , Caenorhabditis elegans Proteins/genetics , Genome , Genomics
4.
Article in English | MEDLINE | ID: mdl-35176489

ABSTRACT

As human life expectancy increases, neurodegenerative diseases present a growing public health threat, for which there are currently few effective treatments. There is an urgent need to understand the molecular and genetic underpinnings of these disorders so new therapeutic targets can be identified. Here we present the argument that the simple nematode worm Caenorhabditis elegans is a powerful tool to rapidly study neurodegenerative disorders due to their short lifespan and vast array of genetic tools, which can be combined with characterization of conserved neuronal processes and behavior orthologous to those disrupted in human disease. We review how pre-existing C. elegans models provide insight into human neurological disease as well as an overview of current tools available to study neurodegenerative diseases in the worm, with an emphasis on genetics and behavior. We also discuss open questions that C. elegans may be particularly well suited for in future studies and how worms will be a valuable preclinical model to better understand these devastating neurological disorders.


Subject(s)
Caenorhabditis elegans , Neurodegenerative Diseases , Animals , Caenorhabditis elegans/genetics , Longevity , Neurodegenerative Diseases/genetics
5.
Mol Psychiatry ; 26(8): 4066-4084, 2021 08.
Article in English | MEDLINE | ID: mdl-33235333

ABSTRACT

Valproate (VPA) has been used in the treatment of bipolar disorder since the 1990s. However, the therapeutic targets of VPA have remained elusive. Here we employ a preclinical model to identify the therapeutic targets of VPA. We find compounds that inhibit histone deacetylase proteins (HDACs) are effective in normalizing manic-like behavior, and that class I HDACs (e.g., HDAC1 and HDAC2) are most important in this response. Using an RNAi approach, we find that HDAC2, but not HDAC1, inhibition in the ventral tegmental area (VTA) is sufficient to normalize behavior. Furthermore, HDAC2 overexpression in the VTA prevents the actions of VPA. We used RNA sequencing in both mice and human induced pluripotent stem cells (iPSCs) derived from bipolar patients to further identify important molecular targets. Together, these studies identify HDAC2 and downstream targets for the development of novel therapeutics for bipolar mania.


Subject(s)
Induced Pluripotent Stem Cells , Valproic Acid , Animals , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Mania , Mice , Valproic Acid/pharmacology
6.
Sci Rep ; 9(1): 20314, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31889133

ABSTRACT

Presynaptic protein synthesis is important in the adult central nervous system; however, the nervous system-wide set of mRNAs localized to presynaptic areas has yet to be identified in any organism. Here we differentially labeled somatic and synaptic compartments in adult C. elegans with fluorescent proteins, and isolated synaptic and somatic regions from the same population of animals. We used this technique to determine the nervous system-wide presynaptic transcriptome by deep sequencing. Analysis of the synaptic transcriptome reveals that synaptic transcripts are predicted to have specialized functions in neurons. Differential expression analysis identified 542 genes enriched in synaptic regions relative to somatic regions, with synaptic functions conserved in higher organisms. We find that mRNAs for pumilio RNA-binding proteins are abundant in synaptic regions, which we confirmed through high-sensitivity in situ hybridization. Presynaptic PUMILIOs regulate associative memory. Our approach enables the identification of new mechanisms that regulate synaptic function and behavior.


Subject(s)
Central Nervous System/physiology , Gene Expression Profiling , Memory , Presynaptic Terminals/metabolism , RNA, Messenger/genetics , Transcriptome , 3' Untranslated Regions , Animals , Binding Sites , Caenorhabditis elegans , Fluorescent Antibody Technique , Gene Expression Regulation , Genome-Wide Association Study , Genomics/methods , Mammals , Neurons/metabolism , Protein Binding , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Synapses/metabolism
7.
Neuron ; 98(3): 562-574.e5, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29656871

ABSTRACT

Perhaps the most devastating decline with age is the loss of memory. Therefore, identifying mechanisms to restore memory function with age is critical. Using C. elegans associative learning and memory assays, we identified a gain-of-function Gαq signaling pathway mutant that forms a long-term (cAMP response element binding protein [CREB]-dependent) memory following one conditioned stimulus-unconditioned stimulus (CS-US) pairing, which usually requires seven CS-US pairings. Increased CREB activity in AIM interneurons reduces the threshold for memory consolidation through transcription of a set of previously identified "long-term memory" genes. Enhanced Gαq signaling in the AWC sensory neuron is both necessary and sufficient for improved memory and increased AIM CREB activity, and activation of Gαq specifically in aged animals rescues the ability to form memory. Activation of Gαq in AWC sensory neurons non-cell autonomously induces consolidation after one CS-US pairing, enabling both cognitive function maintenance with age and restoration of memory function in animals with impaired memory performance without decreased longevity.


Subject(s)
Cognitive Dysfunction/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Memory Consolidation/physiology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Cognitive Dysfunction/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics
8.
Nat Commun ; 8: 14749, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276441

ABSTRACT

Long-lived organisms often feature more stringent protein and DNA quality control. However, whether RNA quality control mechanisms, such as nonsense-mediated mRNA decay (NMD), which degrades both abnormal as well as some normal transcripts, have a role in organismal aging remains unexplored. Here we show that NMD mediates longevity in C. elegans strains with mutations in daf-2/insulin/insulin-like growth factor 1 receptor. We find that daf-2 mutants display enhanced NMD activity and reduced levels of potentially aberrant transcripts. NMD components, including smg-2/UPF1, are required to achieve the longevity of several long-lived mutants, including daf-2 mutant worms. NMD in the nervous system of the animals is particularly important for RNA quality control to promote longevity. Furthermore, we find that downregulation of yars-2/tyrosyl-tRNA synthetase, an NMD target transcript, by daf-2 mutations contributes to longevity. We propose that NMD-mediated RNA surveillance is a crucial quality control process that contributes to longevity conferred by daf-2 mutations.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Longevity/genetics , Mutation , Nonsense Mediated mRNA Decay , RNA/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Gene Expression Profiling , Insulin/genetics , Insulin-Like Growth Factor I/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/genetics
9.
Behav Brain Res ; 322(Pt B): 299-310, 2017 03 30.
Article in English | MEDLINE | ID: mdl-27329151

ABSTRACT

Cognitive decline is a major deficit that arises with age in humans. While some research on the underlying causes of these problems can be done in humans, harnessing the strengths of small model systems, particularly those with well-studied longevity mutants, such as the nematode C. elegans, will accelerate progress. Here we review the approaches being used to study cognitive decline in model organisms and show how simple model systems allow the rapid discovery of conserved molecular mechanisms, which will eventually enable the development of therapeutics to slow cognitive aging.


Subject(s)
Cognitive Aging/physiology , Animals , Caenorhabditis elegans , Cognitive Aging/psychology , Humans , Models, Animal
10.
Curr Biol ; 26(5): 605-15, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26877087

ABSTRACT

Aging is the greatest risk factor for a number of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. Furthermore, normal aging is associated with a decline in sensory, motor, and cognitive functions. Emerging evidence suggests that synapse alterations, rather than neuronal cell death, are the causes of neuronal dysfunctions in normal aging and in early stages of neurodegenerative diseases. However, little is known about the mechanisms underlying age-related synaptic decline. Here, we uncover a surprising role of the anterograde molecular motor UNC-104/KIF1A as a key regulator of neural circuit deterioration in aging C. elegans. Through analyses of synapse protein localization, synaptic transmission, and animal behaviors, we find that reduced function of UNC-104 accelerates motor circuit dysfunction with age, whereas upregulation of UNC-104 significantly improves motor function at advanced ages and also mildly extends lifespan. In addition, UNC-104-overexpressing animals outperform wild-type controls in associative learning and memory tests. Further genetic analyses suggest that UNC-104 functions downstream of the DAF-2-signaling pathway and is regulated by the FOXO transcription factor DAF-16, which contributes to the effects of DAF-2 in neuronal aging. Together, our cellular, electrophysiological, and behavioral analyses highlight the importance of axonal transport in the maintenance of synaptic structural integrity and function during aging and raise the possibility of targeting kinesins to slow age-related neural circuit dysfunction.


Subject(s)
Aging , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , Memory , Nerve Tissue Proteins/genetics , Signal Transduction , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism
11.
Nature ; 529(7584): 92-6, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26675724

ABSTRACT

Insulin/insulin-like growth factor signalling (IIS) is a critical regulator of an organism's most important biological decisions from growth, development, and metabolism to reproduction and longevity. It primarily does so through the activity of the DAF-16 transcription factor (forkhead box O (FOXO) homologue), whose global targets were identified in Caenorhabditis elegans using whole-worm transcriptional analyses more than a decade ago. IIS and FOXO also regulate important neuronal and adult behavioural phenotypes, such as the maintenance of memory and axon regeneration with age, in both mammals and C. elegans, but the neuron-specific IIS/FOXO targets that regulate these functions are still unknown. By isolating adult C. elegans neurons for transcriptional profiling, we identified both the wild-type and IIS/FOXO mutant adult neuronal transcriptomes for the first time. IIS/FOXO neuron-specific targets are distinct from canonical IIS/FOXO-regulated longevity and metabolism targets, and are required for extended memory in IIS daf-2 mutants. The activity of the forkhead transcription factor FKH-9 in neurons is required for the ability of daf-2 mutants to regenerate axons with age, and its activity in non-neuronal tissues is required for the long lifespan of daf-2 mutants. Together, neuron-specific and canonical IIS/FOXO-regulated targets enable the coordinated extension of neuronal activities, metabolism, and longevity under low-insulin signalling conditions.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Forkhead Transcription Factors/metabolism , Insulin/metabolism , Neurons/metabolism , Phenotype , Signal Transduction , Somatomedins/metabolism , Transcriptome , Aging/genetics , Animals , Axons/physiology , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cell Separation , Forkhead Transcription Factors/genetics , Longevity/genetics , Mechanotransduction, Cellular , Memory/physiology , Mutation/genetics , Neurons/cytology , Regeneration/genetics
12.
Neuron ; 85(2): 330-45, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25611510

ABSTRACT

Induced CREB activity is a hallmark of long-term memory, but the full repertoire of CREB transcriptional targets required specifically for memory is not known in any system. To obtain a more complete picture of the mechanisms involved in memory, we combined memory training with genome-wide transcriptional analysis of C. elegans CREB mutants. This approach identified 757 significant CREB/memory-induced targets and confirmed the involvement of known memory genes from other organisms, but also suggested new mechanisms and novel components that may be conserved through mammals. CREB mediates distinct basal and memory transcriptional programs at least partially through spatial restriction of CREB activity: basal targets are regulated primarily in nonneuronal tissues, while memory targets are enriched for neuronal expression, emanating from CREB activity in AIM neurons. This suite of novel memory-associated genes will provide a platform for the discovery of orthologous mammalian long-term memory components.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Developmental/genetics , Hippocampus/metabolism , Memory, Long-Term , Neurons/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Profiling , Gene Expression Regulation/genetics , Learning , Memory , Transcription Factors
13.
Biol Psychiatry ; 77(5): 425-433, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25444159

ABSTRACT

BACKGROUND: Circadian gene disruptions are associated with the development of psychiatric disorders, including addiction. However, the mechanisms by which circadian genes regulate reward remain poorly understood. METHODS: We used mice with a mutation in Npas2 and adeno-associated virus-short hairpin RNA mediated knockdown of Npas2 and Clock in the nucleus accumbens (NAc). We performed conditioned place preference assays. We utilized cell sorting quantitative real-time polymerase chain reaction, and chromatin immunoprecipitation followed by deep sequencing. RESULTS: Npas2 mutants exhibit decreased sensitivity to cocaine reward, which is recapitulated with a knockdown of neuronal PAS domain protein 2 (NPAS2) specifically in the NAc, demonstrating the importance of NPAS2 in this region. Interestingly, reducing circadian locomotor output cycles kaput (CLOCK) (a homologue of NPAS2) in the NAc had no effect, suggesting an important distinction in NPAS2 and CLOCK function. Furthermore, we found that NPAS2 expression is restricted to Drd1 expressing neurons while CLOCK is ubiquitous. Moreover, NPAS2 and CLOCK have distinct temporal patterns of DNA binding, and we identified novel and unique binding sites for each protein. We identified the Drd3 dopamine receptor as a direct transcriptional target of NPAS2 and found that NPAS2 knockdown in the NAc disrupts its diurnal rhythm in expression. Chronic cocaine treatment likewise disrupts the normal rhythm in Npas2 and Drd3 expression in the NAc, which may underlie behavioral plasticity in response to cocaine. CONCLUSIONS: Together, these findings identify an important role for the circadian protein, NPAS2, in the NAc in the regulation of dopamine receptor expression and drug reward.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/drug effects , Receptors, Dopamine D3/metabolism , Reward , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/physiology , Nucleus Accumbens/physiology , Receptors, Dopamine D1/metabolism , Space Perception/drug effects , Space Perception/physiology
14.
Neuropsychopharmacology ; 38(12): 2393-400, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23722243

ABSTRACT

Mice with a mutation in the Clock gene (ClockΔ19) exhibit increased preference for stimulant rewards and sucrose. They also have an increase in dopaminergic activity in the ventral tegmental area (VTA) and a general increase in glutamatergic tone that might underlie these behaviors. However, it is unclear if their phenotype would extend to a very different class of drug (ethanol), and if so, whether these systems might be involved in their response. Continuous access voluntary ethanol intake was evaluated in ClockΔ19 mutants and wild-type (WT) mice. We found that ClockΔ19 mice exhibited significantly increased ethanol intake in a two-bottle choice paradigm. Interestingly, this effect was more robust in female mice. Moreover, chronic ethanol experience resulted in a long-lasting decrease in VTA Clock expression. To determine the importance of VTA Clock expression in ethanol intake, we knocked down Clock expression in the VTA of WT mice via RNA interference. We found that reducing Clock expression in the VTA resulted in significantly increased ethanol intake similar to the ClockΔ19 mice. Interestingly, we also discovered that ClockΔ19 mice exhibit significantly augmented responses to the sedative effects of ethanol and ketamine, but not pentobarbital. However, their drinking behavior was not affected by acamprosate, an FDA-approved drug for the treatment of alcoholism, suggesting that their increased glutamatergic tone might underlie the increased sensitivity to the sedative/hypnotic properties of ethanol but not the rewarding properties of ethanol. Taken together, we have identified a significant role for Clock in the VTA as a negative regulator of ethanol intake and implicate the VTA dopamine system in this response.


Subject(s)
Alcohol Drinking/metabolism , CLOCK Proteins/metabolism , Ethanol/administration & dosage , Ventral Tegmental Area/metabolism , Animals , CLOCK Proteins/genetics , Choice Behavior , Female , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Sex Characteristics
15.
J Neurochem ; 123(1): 124-34, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22757753

ABSTRACT

Mice with a mutation in the Clock gene (ClockΔ19) have a number of behavioral phenotypes that suggest alterations in dopaminergic transmission. These include hyperactivity, increased exploratory behavior, and increased reward value for drugs of abuse. However, the complex changes in dopaminergic transmission that underlie the behavioral abnormalities in these mice remain unclear. Here we find that a loss of CLOCK function increases dopamine release and turnover in striatum as indicated by increased levels of metabolites HVA and DOPAC, and enhances sensitivity to dopamine receptor antagonists. Interestingly, this enlarged dopaminergic tone results in downstream changes in dopamine receptor (DR) levels with a surprising augmentation of both D1- and D2-type DR protein, but a significant shift in the ratio of D1 : D2 receptors in favor of D2 receptor signaling. These effects have functional consequences for both behavior and intracellular signaling, with alterations in locomotor responses to both D1-type and D2-type specific agonists and a blunted response to cAMP activation in the ClockΔ19 mutants. Taken together, these studies further elucidate the abnormalities in dopaminergic transmission that underlie mood, activity, and addictive behaviors.


Subject(s)
CLOCK Proteins/genetics , Gene Expression Regulation/genetics , Mutation/genetics , Receptors, Dopamine/physiology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Colforsin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Homovanillic Acid/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Behav Pharmacol ; 23(4): 392-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22743604

ABSTRACT

Bipolar disorder is a terrible and debilitating disease with limited treatment options. Circadian rhythm disruptions are prominent in bipolar subjects, and studies have shown that rhythm stabilization through psychosocial interventions can improve their symptoms. Furthermore, mice with a mutation in one of the central circadian proteins, CLOCK, have severely disrupted rhythms along with a behavioral profile that closely resembles human mania. A compound has been developed (CK01, similar to PF-670462) that inhibits the activity of casein kinase 1 (CK1), a critical protein involved in the timing of the molecular clock. Previous studies have shown that PF-670462 and other similar compounds are capable of entraining and stabilizing rhythms in arrhythmic animals. Here we show that chronic administration of CK01 leads to a reversal of the anxiety-related behavior, and partial reversal of the depression-related phenotypes of the Clock mutant mouse. This drug had no significant effects on the behavior of wild-type mice at the doses tested. These results suggest that CK1ε/δ inhibitors could be viable drugs for the treatment of bipolar disorder.


Subject(s)
Bipolar Disorder/drug therapy , CLOCK Proteins/genetics , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase Idelta/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Bipolar Disorder/physiopathology , Disease Models, Animal , Drug Administration Schedule , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation
17.
Brain Res ; 1329: 10-20, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20226774

ABSTRACT

Some of the important biochemical, structural, and behavioral changes induced by chronic exposure to drugs of abuse appear to be mediated by the highly stable transcription factor DeltaFosB. Previous work has shown that DeltaFosB overexpression in mice for 2weeks leads to an increase in the expression of numerous genes in striatum, most of which are later downregulated following 8weeks of FosB expression. Interestingly, a large number of these genes were also upregulated in mice overexpressing the transcription factor CREB. It was unclear from this study, however, whether short-term DeltaFosB regulates these genes via CREB. Here, we find that 2weeks of DeltaFosB overexpression increases CREB expression in striatum, an effect that dissipates by 8weeks. The early induction is associated with increased CREB binding to certain target gene promoters in this brain region. Surprisingly, one gene that was a suspected CREB target based on previous reports, cholecystokinin (Cck), was not controlled by CREB in striatum. To further investigate the regulation of Cck following DeltaFosB overexpression, we confirmed that short-term DeltaFosB overexpression increases both Cck promoter activity and gene expression. It also increases binding activity at a putative CREB binding site (CRE) in the Cck promoter. However, while the CRE site is necessary for normal basal expression of Cck, it is not required for DeltaFosB induction of Cck. Taken together, these results suggest that while short-term DeltaFosB induction increases CREB expression and activity at certain gene promoters, this is not the only mechanism by which genes are upregulated under these conditions.


Subject(s)
Cholecystokinin/metabolism , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Animals , Cholecystokinin/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression , Genetic Vectors , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Transgenic , PC12 Cells , Plasmids , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/genetics , Rats , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...