Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biol Psychiatry ; 81(7): 573-584, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27567310

ABSTRACT

BACKGROUND: Addiction relies on persistent alterations of neuronal properties, which depends on gene regulation. Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that modulates neuronal plasticity underlying learning and memory. Its role in cocaine-induced neuronal and behavioral adaptations remains elusive. METHODS: Acute cocaine-treated mice were used for quantitative reverse-transcriptase polymerase chain reaction, immunocytochemistry, and confocal imaging from striatum. Live imaging and transfection assays for Arc overexpression were performed from primary cultures. Molecular and behavioral adaptations to cocaine were studied from Arc-deficient mice and their wild-type littermates. RESULTS: Arc messenger RNA and proteins are rapidly induced in the striatum after acute cocaine administration, via an extracellular-signal regulated kinase-dependent de novo protein synthesis. Although detected in dendrites, Arc accumulates in the nucleus in active zones of transcription, where it colocalizes with phospho-Ser10-histone H3, an important component of nucleosomal response. In vitro, Arc overexpression downregulates phospho-Ser10-histone H3 without modifying extracellular-signal regulated kinase phosphorylation in the nucleus. In vivo, Arc-deficient mice display decreased heterochromatin domains, a high RNA-polymerase II activity and enhanced c-Fos expression. These mice presented an exacerbated psychomotor sensitization and conditioned place preference induced by low doses of cocaine. CONCLUSIONS: Cocaine induces the rapid induction of Arc and its nuclear accumulation in striatal neurons. Locally, it alters the nucleosomal response, and acts as a brake on chromatin remodeling and gene regulation. These original observations posit Arc as a major homeostatic modulator of molecular and behavioral responses to cocaine. Thus, modulating Arc levels may provide promising therapeutic approaches in drug addiction.


Subject(s)
Behavior, Animal/drug effects , Chromatin Assembly and Disassembly , Cocaine/administration & dosage , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Conditioning, Classical/drug effects , Histones/metabolism , Locomotion/drug effects , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , RNA, Messenger
2.
Brain ; 139(Pt 3): 953-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26912634

ABSTRACT

Huntington's disease is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (Exp-HTT) leading to degeneration of striatal neurons. Altered brain cholesterol homeostasis has been implicated in Huntington's disease, with increased accumulation of cholesterol in striatal neurons yet reduced levels of cholesterol metabolic precursors. To elucidate these two seemingly opposing dysregulations, we investigated the expression of cholesterol 24-hydroxylase (CYP46A1), the neuronal-specific and rate-limiting enzyme for cholesterol conversion to 24S-hydroxycholesterol (24S-OHC). CYP46A1 protein levels were decreased in the putamen, but not cerebral cortex samples, of post-mortem Huntington's disease patients when compared to controls. Cyp46A1 mRNA and CYP46A1 protein levels were also decreased in the striatum of the R6/2 Huntington's disease mouse model and in SThdhQ111 cell lines. In vivo, in a wild-type context, knocking down CYP46A1 expression in the striatum, via an adeno-associated virus-mediated delivery of selective shCYP46A1, reproduced the Huntington's disease phenotype, with spontaneous striatal neuron degeneration and motor deficits, as assessed by rotarod. In vitro, CYP46A1 restoration protected SThdhQ111 and Exp-HTT-expressing striatal neurons in culture from cell death. In the R6/2 Huntington's disease mouse model, adeno-associated virus-mediated delivery of CYP46A1 into the striatum decreased neuronal atrophy, decreased the number, intensity level and size of Exp-HTT aggregates and improved motor deficits, as assessed by rotarod and clasping behavioural tests. Adeno-associated virus-CYP46A1 infection in R6/2 mice also restored levels of cholesterol and lanosterol and increased levels of desmosterol. In vitro, lanosterol and desmosterol were found to protect striatal neurons expressing Exp-HTT from death. We conclude that restoring CYP46A1 activity in the striatum promises a new therapeutic approach in Huntington's disease.


Subject(s)
Cholesterol/metabolism , Huntington Disease/enzymology , Huntington Disease/prevention & control , Steroid Hydroxylases/biosynthesis , Aged , Aged, 80 and over , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cholesterol 24-Hydroxylase , Female , Humans , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Middle Aged
3.
J Neurosci ; 31(40): 14296-307, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976515

ABSTRACT

Activation of the extracellular signal-regulated kinase (ERK) signaling pathway in the striatum is crucial for molecular adaptations and long-term behavioral alterations induced by cocaine. In response to cocaine, ERK controls the phosphorylation levels of both mitogen and stress-activated protein kinase 1 (MSK-1), a nuclear kinase involved in histone H3 (Ser10) and cAMP response element binding protein phosphorylation, and Elk-1, a transcription factor involved in serum response element (SRE)-driven gene regulations. We recently characterized the phenotype of msk-1 knock-out mice in response to cocaine. Herein, we wanted to address the role of Elk-1 phosphorylation in cocaine-induced molecular, morphological, and behavioral responses. We used a cell-penetrating peptide, named TAT-DEF-Elk-1 (TDE), which corresponds to the DEF docking domain of Elk-1 toward ERK and inhibits Elk-1 phosphorylation induced by ERKs without modifying ERK or MSK-1 in vitro. The peptide was injected in vivo before cocaine administration in mice. Immunocytochemical, molecular, morphological, and behavioral studies were performed. The TDE inhibited Elk-1 and H3 (Ser10) phosphorylation induced by cocaine, sparing ERK and MSK-1 activation. Consequently, TDE altered cocaine-induced regulation of genes bearing SRE site(s) in their promoters, including c-fos, zif268, ΔFosB, and arc/arg3.1 (activity-regulated cytoskeleton-associated protein). In a chronic cocaine administration paradigm, TDE reversed cocaine-induced increase in dendritic spine density. Finally, the TDE delayed the establishment of cocaine-induced psychomotor sensitization and conditioned-place preference. We conclude that Elk-1 phosphorylation downstream from ERK is a key molecular event involved in long-term neuronal and behavioral adaptations to cocaine.


Subject(s)
Cocaine/pharmacology , Neural Inhibition/physiology , ets-Domain Protein Elk-1/antagonists & inhibitors , ets-Domain Protein Elk-1/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neural Inhibition/drug effects , Peptides/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology
4.
Hum Mol Genet ; 20(12): 2422-34, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21493629

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder due to abnormal polyglutamine expansion in huntingtin protein (Exp-Htt). This expansion causes protein aggregation, leading to neuronal dysfunction and death. We have previously shown that mitogen- and stress-activated kinase (MSK-1), a nuclear protein kinase involved in chromatin remodeling through histone H3 phosphorylation, is deficient in the striatum of HD patients and model mice. Restoring MSK-1 expression in cultured striatal cells prevented neuronal dysfunction and death induced by Exp-Htt. Here we extend these observations in a rat model of HD based on striatal lentiviral expression of Exp-Htt (LV-Exp-HTT). MSK-1 overexpression attenuated Exp-Htt-induced down-regulation of DARPP-32 expression 4 and 10 weeks after infection and enhanced NeuN staining after 10 weeks. LV-MSK-1 induced constitutive hyperphosphorylation of H3 and cAMP-responsive element binding protein (CREB), indicating that MSK-1 has spontaneous catalytic activity. MSK-1 overexpression also upregulated peroxisome proliferator-activated receptor γ coactivator alpha (PGC-1α), a transcriptional co-activator involved in mitochondrial biogenesis. Chromatin immunoprecipitation indicated that transcriptional regulation of PGC-1α is directly linked to increased binding of MSK-1, along with H3 and CREB phosphorylation of the PGC-1α promoter. MSK-1 knock-out mice showed spontaneous striatal atrophy as they aged, as well as higher susceptibility to systemic administration of the mitochondrial neurotoxin 3-NP. These results indicate that MSK-1 activation is an important and key event in the signaling cascade that regulates PGC-1α expression. Strategies aimed at restoring MSK-1 expression in the striatum might offer a new therapeutic approach to HD.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Corpus Striatum/metabolism , Gene Expression Regulation/drug effects , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Nuclear Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/pharmacology , Analysis of Variance , Animals , Chromatin Assembly and Disassembly/physiology , Chromatin Immunoprecipitation , DNA Repeat Expansion/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation/physiology , Genetic Vectors/genetics , Huntingtin Protein , Immunohistochemistry , Lentivirus , Mice , Mice, Knockout , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Neuroprotective Agents/metabolism , Nuclear Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Biol Psychiatry ; 69(3): 218-27, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21055728

ABSTRACT

BACKGROUND: Activation of the extracellular signal-regulated kinase (ERK) in the striatum is crucial for long-term behavioral alterations induced by drugs of abuse. In response to cocaine, ERK phosphorylation (i.e., activation) is restricted to medium-sized spiny neurons expressing dopamine D1 receptor (D1R) and depends on a concomitant stimulation of D1R and glutamate N-methyl-D-aspartate receptor (NMDAR). However, the mechanisms responsible for this activation, especially the respective contribution of D1R and NMDAR, remain unknown. METHODS: We studied striatal neurons in culture stimulated with D1R agonist and/or glutamate and wild-type or genetically modified mice treated with cocaine. Biochemical, immunohistochemical, and imaging studies were performed. Mice were also subjected to behavioral experiments. RESULTS: Stimulation of D1R cannot activate ERK by itself but potentiates glutamate-mediated calcium influx through NMDAR that is responsible for ERK activation. Potentiation of NMDAR by D1R depends on a cyclic adenosine monophosphate-independent signaling pathway, which involves tyrosine phosphorylation of the NR2B subunit of NMDAR by Src family kinases. We also demonstrate that the D1R/Src family kinases/NR2B pathway is responsible for ERK activation by cocaine in vivo. Inhibition of this pathway abrogates cocaine-induced locomotor sensitization and conditioned place preference. CONCLUSIONS: Our results show that potentiation of NR2B-containing NMDAR by D1R is necessary and sufficient to trigger cocaine-induced ERK activation. They highlight a new cyclic adenosine monophosphate-independent pathway responsible for the integration of dopamine and glutamate signals by the ERK cascade in the striatum and for long-term behavioral alterations induced by cocaine.


Subject(s)
Cocaine/pharmacology , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Tyrosine/metabolism , Animals , Calcium/metabolism , Conditioning, Psychological/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Drug Interactions , Glutamic Acid/pharmacology , Mice , Motor Activity/drug effects , Neurons/metabolism , Phenols/pharmacology , Phosphorylation/physiology , Piperidines/pharmacology , Receptors, Dopamine D1/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , src-Family Kinases/metabolism
6.
Mol Cell Neurosci ; 41(3): 325-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398002

ABSTRACT

Nuclear translocation of activated extracellular signal-regulated kinases (ERK) in neurons is critical for gene regulations underlying long-term neuronal adaptation and memory formation. However, it is unknown how activated ERK travel from the post-synaptic elements where their activation occurs, to the nucleus where they translocate to exert their transcriptional roles. In cultured neurons, we identified endocytosis as a prime event in glutamate-induced nuclear trafficking of ERK2. We show that glutamate triggers a rapid recruitment of ERK2 to a protein complex comprising markers of the clathrin-dependent endocytotic and AMPA/glutamate receptor subtype. Inhibition of endocytosis results in a neuritic withholding of activated ERK2 without modification of ERK2 activity. As a consequence, endocytosis blockade alters ERK-dependent nuclear events, such as mitogen and stressed-activated kinase-1 (MSK-1) activation, histone H3 phosphorylation and gene regulations. Our data provide the first evidence that the endocytic pathway controls ERK nuclear translocation and ERK-dependent gene regulations induced by glutamate.


Subject(s)
Cell Nucleus/metabolism , Endocytosis/drug effects , Glutamic Acid/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Animals , Cell Culture Techniques , Cell Line , Cell Nucleus/drug effects , Chlorocebus aethiops , Chromatin Assembly and Disassembly , Enzyme Activation , Neurons/drug effects , Neurons/metabolism , Protein Transport/drug effects , Rats , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , ets-Domain Protein Elk-1/metabolism
7.
FASEB J ; 22(4): 1083-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18029446

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder due to an abnormal polyglutamine expansion in the N-terminal region of huntingtin protein (Exp-Htt). This expansion causes protein aggregation and neuronal dysfunction and death. Transcriptional dysregulation due to Exp-Htt participates in neuronal death in HD. Here, using the R6/2 transgenic mouse model of HD, we identified a new molecular alteration that could account for gene dysregulation in these mice. Despite a nuclear activation of the mitogen-activated protein kinase/extracellular regulated kinase (ERK) along with Elk-1 and cAMP responsive element binding, two transcription factors involved in c-Fos transcription, we failed to detect any histone H3 phosphorylation, which is expected after nuclear ERK activation. Accordingly, we found in the striatum of these mice a deficiency of mitogen- and stress-activated kinase-1 (MSK-1), a kinase downstream ERK, critically involved in H3 phosphorylation and c-Fos induction. We extended this observation to Exp-Htt-expressing striatal neurons and postmortem brains of HD patients. In vitro, knocking out MSK-1 expression potentiated Exp-Htt-induced striatal death. Its overexpression induced H3 phosphorylation and c-Fos expression and totally protected against striatal neurodegeneration induced by Exp-Htt. We propose that MSK-1 deficiency is involved in transcriptional dysregulation and striatal degeneration. Restoration of its expression and activity may be a new therapeutic target in HD.


Subject(s)
Corpus Striatum/enzymology , DNA Repeat Expansion , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Transcription, Genetic , Animals , Corpus Striatum/metabolism , Down-Regulation , Genes, fos , Histones/metabolism , Huntingtin Protein , Huntington Disease/enzymology , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics
8.
Neurobiol Dis ; 29(1): 22-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17905594

ABSTRACT

Huntington's disease (HD) results from an abnormal polyglutamine extension in the N-terminal region of the huntingtin protein. This mutation causes preferential degeneration of striatal projection neurons. We previously demonstrated, in vitro, that dopaminergic D2 receptor stimulation acted synergistically with mutated huntingtin (expHtt) to increase aggregate formation and striatal death. In the present work, we extend these observations to an in vivo system based on lentiviral-mediated expression of expHtt in the rat striatum. The early and chronic treatment with the D2 antagonist haloperidol decanoate protects striatal neurons from expHtt-induced dysfunction, as analyzed by DARPP-32 and NeuN stainings. Haloperidol treatment also reduces aggregates formation, an effect that is maintained over time. These findings indicate that D2 receptors activation contributes to the deleterious effects of expHtt on striatal function and may represent an interesting early target to alter the subsequent course of neuropathology in HD.


Subject(s)
Antipsychotic Agents/therapeutic use , Corpus Striatum/pathology , Haloperidol/analogs & derivatives , Huntington Disease/prevention & control , Mutation/physiology , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nuclear Proteins/genetics , Age Factors , Animals , Cell Count/methods , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Female , Gene Expression Regulation/drug effects , Haloperidol/therapeutic use , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Lentivirus/physiology , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar
9.
J Neurosci ; 27(52): 14448-58, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18160653

ABSTRACT

The transcription factor Elk-1 plays a key role in cell differentiation, proliferation and apoptosis. This role is thought to arise from its phosphorylation by activated extracellular signal-regulated kinases (ERKs), a critical posttranslational event for the transcriptional activity of the ternary complex composed of Elk-1 and a dimer of serum response factor (SRF) at the serum response element (SRE) regulatory site of transcription. In addition to its nuclear localization, Elk-1 is found in the dendrites and soma of neuronal cells and recent evidence implicate a cytoplasmic proapoptotic function of Elk-1, via its association with the mitochondrial permeability transition pore complex. Thus, the nuclear versus cytoplasmic localization of Elk-1 seems to be crucial for its biological function. In this study we show that the excitatory neurotransmitter, glutamate, induces an ERK-dependent Elk-1 activation and nuclear relocalization. We demonstrate that Elk-1 phosphorylation on Ser383/389 has a dual function and triggers both Elk-1 nuclear translocation and SRE-dependent gene expression. Mutating these sites into inactive residues or using a synthetic penetrating peptide (TAT-DEF-Elk-1), which specifically interferes with the DEF docking domain of Elk-1, prevents Elk-1 nuclear translocation without interfering with ERK nor MSK1 (mitogen- and stress-activated protein kinase 1), a CREB kinase downstream from ERK- activation. This results in a differential regulation of glutamate-induced IEG regulation when compared with classical inhibitors of the ERK pathway. Using the TAT-DEF-Elk-1 peptide or the dominant-negative version of Elk-1, we show that Elk-1 phosphorylation controls dendritic elongation, SRF and Actin expression levels as well as cytoskeleton dynamics.


Subject(s)
Cytoskeleton/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Peptides/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cocaine/pharmacology , Corpus Striatum/cytology , Dopamine Uptake Inhibitors/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Peptides/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Serine/metabolism , Serine/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transfection/methods , Transfection/statistics & numerical data
10.
J Neurochem ; 101(3): 697-708, 2007 May.
Article in English | MEDLINE | ID: mdl-17241117

ABSTRACT

Chromatin remodelling is thought to play a key role in gene regulation that underlies long-term synaptic plasticity and memory formation. The dynamic process of chromatin remodelling requires post-translational modifications of histones, a group of highly basic proteins that are tightly linked to DNA. In the present study, we investigated histone H3 modifications in response to glutamate stimulation leading to c-Fos and c-Jun induction in an in vitro model system of striatal neurons in culture. Intracellular signalling pathways implicated in these modifications were analysed. Histone H3 acetylation was strong in basal conditions and unmodified by glutamate treatment. By contrast, glutamate induced a strong phosphorylation of histone H3 that was inhibited by selective inhibitors of the extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38 MAPK) pathways, U0126 and SB203580, respectively. Blocking activation of mitogen- and stress-activated kinase 1 (MSK1), a kinase downstream ERK and p38 MAPK, by pharmacological approach or using striatal cells from MSK1 deficient mice, totally abolished H3 phosphorylation, as well as c-Fos and c-Jun induction. Chromatin immunoprecipitation assays confirmed increased levels of phosphorylated H3 at the c-jun promoter. Altogether, our data highlight the crucial role of MSK1 in the nucleosomal response necessary for gene induction in neuronal cells.


Subject(s)
Corpus Striatum/cytology , Glutamic Acid/pharmacology , Histones/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 8/physiology , Neurons/drug effects , Acetylation/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 8/deficiency , Models, Neurological , Phosphorylation/drug effects , Transcriptional Activation
11.
J Neurosci ; 25(49): 11444-54, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16339038

ABSTRACT

Although the induction of persistent behavioral alterations by drugs of abuse requires the regulation of gene transcription, the precise intracellular signaling pathways that are involved remain mainly unknown. Extracellular signal-regulated kinase (ERK) is critical for the expression of immediate-early genes in the striatum in response to cocaine and Delta9-tetrahydrocannabinol and for the rewarding properties of these drugs. Here we show that in mice a single injection of cocaine (10 mg/kg) activates mitogen- and stress-activated protein kinase 1 (MSK1) in dorsal striatum and nucleus accumbens. Cocaine-induced phosphorylation of MSK1 threonine 581 and cAMP response element-binding protein (CREB) serine 133 (Ser133) were blocked by SL327, a drug that prevents ERK activation. Cocaine increased the acetylation of histone H4 lysine 5 and phosphorylation of histone H3 Ser10, demonstrating the existence of drug-induced chromatin remodeling in vivo. In MSK1 knock-out (KO) mice CREB and H3 phosphorylation in response to cocaine (10 mg/kg) were blocked, and induction of c-Fos and dynorphin was prevented, whereas the induction of Egr-1 (early growth response-1)/zif268/Krox24 was unaltered. MSK1-KO mice had no obvious neurological defect but displayed a contrasted behavioral phenotype in response to cocaine. Acute effects of cocaine and dopamine D1 or D2 agonists were unaltered. Sensitivity to low doses, but not high doses, of cocaine was increased in the conditioned place preference paradigm, whereas locomotor sensitization to repeated injections of cocaine was decreased markedly. Our results show that MSK1 is a major striatal kinase, downstream from ERK, responsible for the phosphorylation of CREB and H3 and is required specifically for the induction of c-Fos and dynorphin as well as for locomotor sensitization.


Subject(s)
Cocaine/pharmacology , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/metabolism , Motor Activity/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 8/genetics , Motor Activity/genetics
12.
Proc Natl Acad Sci U S A ; 102(34): 12218-23, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16103364

ABSTRACT

Huntington's disease (HD), an inherited neurodegenerative disorder, results from an abnormal polyglutamine extension in the N-terminal region of the huntingtin protein. This mutation leads to protein aggregation and neurotoxicity. Despite its widespread expression in the brain and body, mutated huntingtin causes selective degeneration of striatal projection neurons. In the present study, we investigate the role of dopamine (DA) in this preferential vulnerability. Using primary cultures of striatal neurons transiently expressing GFP-tagged-exon 1 of mutated huntingtin, we show that low doses of DA (100 microM) act synergistically with mutated huntingtin to activate the proapoptotic transcription factor c-Jun. Surprisingly, DA also increases aggregate formation of mutated huntingtin in all cellular compartments, including neurites, soma, and nuclei. DA-dependent potentiation of c-Jun activation was reversed by ascorbate, a reactive oxygen species (ROS) scavenger, and SP-600125, a selective inhibitor of the c-Jun N-terminal kinase (JNK) pathway. By contrast, DA effects on aggregate formation were reversed by a selective D2 receptor antagonist and reproduced by a D2 agonist. Similarly, striatal neurons from D2 knockout mice showed no effect of DA on aggregate formation. Blocking ROS production, JNK activation, or D2 receptor stimulation significantly reversed DA aggravation of mutated huntingtin-induced striatal death. The combined treatment with the ROS scavenger and D2 antagonist totally reversed DA's effects on mutated huntingtin-induced striatal death. Thus, the present results provide insights into the cellular mechanisms that govern striatal vulnerability in HD and strongly support a dual role of JNK activation and D2 receptor signaling in this process.


Subject(s)
Dopamine/pharmacology , Gene Expression Regulation/drug effects , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Analysis of Variance , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Dopamine/metabolism , Dopamine D2 Receptor Antagonists , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Dopamine D2/agonists , Signal Transduction/physiology
13.
Eur J Neurosci ; 19(7): 1826-36, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15078556

ABSTRACT

A major goal of research on addiction is to identify the molecular mechanisms of long-lasting behavioural alterations induced by drugs of abuse. Cocaine and delta-9-tetrahydrocannabinol (THC) activate extracellular signal-regulated kinase (ERK) in the striatum and blockade of the ERK pathway prevents establishment of conditioned place preference to these drugs. However, it is not known whether activation of ERK in the striatum is specific for these two drugs and/or this brain region. We studied the appearance of phospho-ERK immunoreactive neurons in CD-1 mouse brain following acute administration of drugs commonly abused by humans, cocaine, morphine, nicotine and THC, or of other psychoactive compounds including caffeine, scopolamine, antidepressants and antipsychotics. Each drug generated a distinct regional pattern of ERK activation. All drugs of abuse increased ERK phosphorylation in nucleus accumbens, lateral bed nucleus of the stria terminalis, central amygdala and deep layers of prefrontal cortex, through a dopamine D1 receptor-dependent mechanism. Although some non-addictive drugs moderately activated ERK in a few of these areas, they never induced this combined pattern of strong activation. Antidepressants and caffeine activated ERK in hippocampus and cerebral cortex. Typical antipsychotics mildly activated ERK in dorsal striatum and superficial prefrontal cortex, whereas clozapine had no effect in the striatum, but more widespread effects in cortex and amygdala. Our results outline a subset of structures in which ERK activation might specifically contribute to the long-term effects of drugs of abuse, and suggest mapping ERK activation in brain as a way to identify potential sites of action of psychoactive drugs.


Subject(s)
Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/pharmacology , Brain/cytology , Cocaine/pharmacology , Dronabinol/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Benzazepines/pharmacology , Brain/drug effects , Brain/metabolism , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Desipramine/pharmacology , Dopamine Antagonists/pharmacology , Enzyme Activation/drug effects , Fluoxetine/pharmacology , Immunohistochemistry/methods , Male , Mice , Morphine/pharmacology , Muscarinic Antagonists/pharmacology , Neurons/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Scopolamine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
14.
J Neurosci ; 22(20): 8911-21, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12388598

ABSTRACT

Akt is classically described as a prosurvival serine/threonine kinase activated in response to trophic factors. After activation by phosphoinositide 3-kinase (PI3-kinase), it can translocate to the nucleus where it promotes specific genetic programs by catalyzing phosphorylation of transcription factors. We report here that both dopamine (DA) D1 (SKF38393) and D2 (quinpirole) agonist treatments rapidly increase, in primary striatal neurons in culture, phosphorylation levels of Akt on Thr(308), a residue that is critically involved in its kinase activity. These treatments also activate the extracellular signal-regulated kinase (ERK) pathway in the same population of striatal neurons. Induction of active, phospho-Thr(308) Akt by dopamine D1 and D2 agonists is insensitive to wortmannin and thus PI3-kinase independent, in contrast to growth factor-induced Akt activity. D1- and D2-induced phospho-Thr(308) Akt is decreased by the mitogen-activated protein kinase kinase (MEK) inhibitor, U0126, as well as by overexpression of a dominant-negative version of MEK, thus implicating the Ras/ERK signaling cascade in this process. Furthermore, overexpression of a mutant form of Akt that cannot be activated impaired cAMP response element-binding protein (CREB) phosphorylation induced by SKF38393 and quinpirole treatments. Activation of Akt on Thr(308) was also found in vivo in striatal neurons after acute administration of cocaine, a psychostimulant that strongly increases DA transmission. Thus, multiple intracellular pathways can transduce signals from dopamine receptors to CREB in striatal neurons, one of these being Akt. We propose that this signaling pathway plays a pivotal role in DA-induced regulation of gene expression and long-term neuronal adaptation in the striatum.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Dopamine/pharmacology , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Cells, Cultured , Cocaine/pharmacology , Corpus Striatum/cytology , Enzyme Activation/drug effects , Genes, Dominant , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Phosphorylation/drug effects , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Subcellular Fractions/chemistry , Transfection
15.
J Neurosci ; 22(6): 2174-84, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11896157

ABSTRACT

Impairments in mitochondrial energy metabolism are thought to be involved in most neurodegenerative diseases, including Huntington's disease (HD). Chronic administration of 3-nitropropionic acid (3-NP), a suicide inhibitor of succinate dehydrogenase, causes prolonged energy impairments and replicates most of the pathophysiological features of HD, including preferential striatal degeneration. In this study, we analyzed one of the mechanisms that could account for this selective 3-NP-induced striatal degeneration. In chronically 3-NP-infused rats, the time course of motor behavioral impairments and histological abnormalities was determined. Progressive alterations of motor performance occurred after 3 d. By histological analysis and terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end-labeling staining, we found a selective neurodegenerescence in the striatum, occurring first in its dorsolateral (DL) part. Activation of c-Jun N-terminal kinase (JNK) was analyzed from brain sections of these rats, using immunocytochemical detection of its phosphorylated form. Activation of JNK occurred progressively and selectively in the DL of the striatum and was followed by c-Jun activation and expression in the same striatal region. To elucidate the role of the JNK/c-Jun module in 3-NP-induced striatal degeneration, we then used primary striatal neurons in culture, in which we replicated neuronal death by application of 3-NP. We found strong nuclear translocation of activated JNK that was rapidly followed by phosphorylation of the transcription factor c-Jun. Overexpression of a dominant negative version of c-Jun, lacking its transactivation domain and phosphorylation sites for activated JNK, completely abolished 3-NP-induced striatal neurodegeneration. We thus conclude that a genetic program controlled by the JNK/c-Jun module is an important molecular event in 3-NP-induced striatal degeneration.


Subject(s)
Corpus Striatum/drug effects , Huntington Disease/metabolism , Mitogen-Activated Protein Kinases/metabolism , Propionates/toxicity , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/pathology , Disease Models, Animal , Disease Progression , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, Dominant , Huntington Disease/chemically induced , Huntington Disease/pathology , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases , Male , Mutagenesis, Site-Directed , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitro Compounds , Phosphorylation/drug effects , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...