Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 11(4): e0153327, 2016.
Article in English | MEDLINE | ID: mdl-27065006

ABSTRACT

Cannabis is the most widely used illicit drug in the world. Delta-9-tetrahydrocannabinol (Δ9-THC) is the main psychoactive component of cannabis and its effects have been well-studied. However, cannabis contains many other cannabinoids that affect brain function. Therefore, these studies investigated the effect of cannabis smoke exposure on locomotor activity, rearing, anxiety-like behavior, and the development of dependence in rats. It was also investigated if cannabis smoke exposure leads to tolerance to the locomotor-suppressant effects of the endogenous cannabinoid anandamide. Cannabis smoke was generated by burning 5.7% Δ9-THC cannabis cigarettes in a smoking machine. The effect of cannabis smoke on the behavior of rats in a small and large open field and an elevated plus maze was evaluated. Cannabis smoke exposure induced a brief increase in locomotor activity followed by a prolonged decrease in locomotor activity and rearing in the 30-min small open field test. The cannabinoid receptor type 1 (CB1) receptor antagonist rimonabant increased locomotor activity and prevented the smoke-induced decrease in rearing. Smoke exposure also increased locomotor activity in the 5-min large open field test and the elevated plus maze test. The smoke exposed rats spent more time in the center zone of the large open field, which is indicative of a decrease in anxiety-like behavior. A high dose of anandamide decreased locomotor activity and rearing in the small open field and this was not prevented by rimonabant or pre-exposure to cannabis smoke. Serum Δ9-THC levels were 225 ng/ml after smoke exposure, which is similar to levels in humans after smoking cannabis. Exposure to cannabis smoke led to dependence as indicated by more rimonabant-precipitated somatic withdrawal signs in the cannabis smoke exposed rats than in the air-control rats. In conclusion, chronic cannabis smoke exposure in rats leads to clinically relevant Δ9-THC levels, dependence, and has a biphasic effect on locomotor activity.


Subject(s)
Arachidonic Acids/pharmacology , Behavior, Animal/drug effects , Endocannabinoids/pharmacology , Marijuana Smoking/physiopathology , Polyunsaturated Alkamides/pharmacology , Animals , Drug Tolerance , Exploratory Behavior/drug effects , Male , Marijuana Abuse/etiology , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Wistar , Time Factors
2.
J Neurochem ; 137(2): 164-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26826701

ABSTRACT

Neuronal activity sculpts brain development by inducing the transcription of genes such as brain-derived neurotrophic factor (Bdnf) that modulate the function of synapses. Sensory experience is transduced into changes in gene transcription via the activation of calcium signaling pathways downstream of both L-type voltage-gated calcium channels (L-VGCCs) and NMDA-type glutamate receptors (NMDARs). These signaling pathways converge on the regulation of transcription factors including calcium-response factor (CaRF). Although CaRF is dispensable for the transcriptional induction of Bdnf following the activation of L-VGCCs, here we show that the loss of CaRF leads to enhanced NMDAR-dependent transcription of Bdnf as well as Arc. We identify the NMDAR subunit-encoding gene Grin3a as a regulatory target of CaRF, and we show that expression of both Carf and Grin3a is depressed by the elevation of intracellular calcium, linking the function of this transcriptional regulatory pathway to neuronal activity. We find that light-dependent activation of Bdnf and Arc transcription is enhanced in the visual cortex of young CaRF knockout mice, suggesting a role for CaRF-dependent dampening of NMDAR-dependent transcription in the developing brain. Finally, we demonstrate that enhanced Bdnf expression in CaRF-lacking neurons increases inhibitory synapse formation. Taken together, these data reveal a novel role for CaRF as an upstream regulator of NMDAR-dependent gene transcription and synapse formation in the developing brain. NMDARs promote brain development by inducing the transcription of genes, including brain-derived neurotrophic factor (BDNF). We show that the transcription factor calcium-response factor (CaRF) limits NMDAR-dependent BDNF induction by regulating expression of the NMDAR subunit GluN3A. Loss of CaRF leads to enhanced BDNF-dependent GABAergic synapse formation indicating the importance of this process for brain development. Our observation that both CaRF and GluN3A are down-regulated by intracellular calcium suggests that this may be a mechanism for experience-dependent modulation of synapse formation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/growth & development , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/physiology , Membrane Glycoproteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/genetics , Calcium Channel Blockers/pharmacology , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Tetrodotoxin/pharmacology , Transcription Factors/genetics , Valine/analogs & derivatives , Valine/pharmacology , Visual Cortex/metabolism
3.
J Neurosci ; 34(13): 4519-27, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24671997

ABSTRACT

The methyl-DNA binding protein MeCP2 is emerging as an important regulator of drug reinforcement processes. Psychostimulants induce phosphorylation of MeCP2 at Ser421; however, the functional significance of this posttranslational modification for addictive-like behaviors was unknown. Here we show that MeCP2 Ser421Ala knock-in mice display both a reduced threshold for the induction of locomotor sensitization by investigator-administered amphetamine and enhanced behavioral sensitivity to the reinforcing properties of self-administered cocaine. These behavioral differences were accompanied in the knock-in mice by changes in medium spiny neuron intrinsic excitability and nucleus accumbens gene expression typically observed in association with repeated exposure to these drugs. These data show that phosphorylation of MeCP2 at Ser421 functions to limit the circuit plasticities in the nucleus accumbens that underlie addictive-like behaviors.


Subject(s)
Central Nervous System Stimulants/pharmacology , Exploratory Behavior/drug effects , Methyl-CpG-Binding Protein 2/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Amphetamine/pharmacology , Animals , Cocaine/administration & dosage , Corpus Striatum/cytology , Dopamine Uptake Inhibitors/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Phosphorylation/physiology , Self Administration
4.
Mol Ther ; 21(1): 18-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23011033

ABSTRACT

Typical Rett syndrome (RTT) is a pediatric disorder caused by loss-of-function mutations in the methyl-CpG binding protein 2 (MECP2) gene. The demonstrated reversibility of RTT-like phenotypes in mice suggests that MECP2 gene replacement is a potential therapeutic option in patients. We report improvements in survival and phenotypic severity in Mecp2-null male mice after neonatal intracranial delivery of a single-stranded (ss) AAV9/chicken ß-actin (CBA)-MECP2 vector. Median survival was 16.6 weeks for MECP2-treated versus 9.3 weeks for green fluorescent protein (GFP)-treated mice. ssAAV9/CBA-MECP2-treated mice also showed significant improvement in the phenotype severity score, in locomotor function, and in exploratory activity, as well as a normalization of neuronal nuclear volume in transduced cells. Wild-type (WT) mice receiving neonatal injections of the same ssAAV9/CBA-MECP2 vector did not show any significant deficits, suggesting a tolerance for modest MeCP2 overexpression. To test a MECP2 gene replacement approach in a manner more relevant for human translation, a self-complementary (sc) adeno-associated virus (AAV) vector designed to drive MeCP2 expression from a fragment of the Mecp2 promoter was injected intravenously (IV) into juvenile (4-5 weeks old) Mecp2-null mice. While the brain transduction efficiency in juvenile mice was low (~2-4% of neurons), modest improvements in survival were still observed. These results support the concept of MECP2 gene therapy for RTT.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy , Mice, Knockout/physiology , Rett Syndrome/therapy , Survival Rate , Animals , Animals, Newborn , Brain/metabolism , Male , Mice , Mice, Knockout/genetics , Phenotype , Rett Syndrome/genetics
5.
Neuropsychopharmacology ; 37(2): 321-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21956448

ABSTRACT

Systemic administration of amphetamine (AMPH) induces phosphorylation of MeCP2 at Ser421 (pMeCP2) in select populations of neurons in the mesolimbocortical brain regions. Because AMPH simultaneously activates multiple monoamine neurotransmitter systems, here we examined the ability of dopamine (DA), serotonin (5-HT), and norepinephrine (NE) to induce pMeCP2. Selective blockade of the DA transporter (DAT) or the 5-HT transporter (SERT), but not the NE transporter (NET), was sufficient to induce pMeCP2 in the CNS. DAT blockade induced pMeCP2 in the prelimbic cortex (PLC) and nucleus accumbens (NAc), whereas SERT blockade induced pMeCP2 only in the NAc. Administration of selective DA and 5-HT receptor agonists was also sufficient to induce pMeCP2; however, the specific combination of DA and 5-HT receptors activated determined the regional- and cell-type specificity of pMeCP2 induction. The D(1)-class DA receptor agonist SKF81297 induced pMeCP2 widely; however, coadministration of the D(2)-class agonist quinpirole restricted the induction of pMeCP2 to GABAergic interneurons of the NAc. Intra-striatal injection of the adenylate cyclase activator forskolin was sufficient to induce pMeCP2 in medium-spiny neurons, suggesting that the combinatorial regulation of cAMP by different classes of DA and 5-HT receptors may contribute to the cell-type specificity of pMeCP2 induction. Consistent with the regulation of pMeCP2 by multiple monoamine neurotransmitters, genetic disruption of any single monoamine transporter in DAT-, SERT-, and NET-knockout mice failed to eliminate AMPH-induced pMeCP2 in the NAc. Together, these studies indicate that combinatorial signaling through DA and 5-HT receptors can regulate the brain region- and cell-type specific pMeCP2 in the CNS.


Subject(s)
Brain/metabolism , Dopamine/physiology , Gene Expression Regulation/physiology , Methyl-CpG-Binding Protein 2/metabolism , Serotonin/physiology , Animals , Brain/drug effects , Cells, Cultured , Citalopram/pharmacology , Colforsin/pharmacology , Dopamine Agonists/pharmacology , Drug Interactions , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Gene Expression Regulation/drug effects , Male , Mice , Mice, Knockout , Microinjections , Molecular Imaging/methods , Morpholines/pharmacology , Motor Activity/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Piperazines/pharmacology , Plasma Membrane Neurotransmitter Transport Proteins/antagonists & inhibitors , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Quipazine/pharmacology , Reboxetine
6.
Nat Neurosci ; 13(9): 1128-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20711186

ABSTRACT

MeCP2 is a methyl DNA-binding transcriptional regulator that contributes to the development and function of CNS synapses; however, the requirement for MeCP2 in stimulus-regulated behavioral plasticity is not fully understood. Here we show that acute viral manipulation of MeCP2 expression in the nucleus accumbens (NAc) bidirectionally modulates amphetamine (AMPH)-induced conditioned place preference. Mecp2 hypomorphic mutant mice have more NAc GABAergic synapses and show deficient AMPH-induced structural plasticity of NAc dendritic spines. Furthermore, these mice show deficient plasticity of striatal immediate early gene inducibility after repeated AMPH administration. Notably, psychostimulants induce phosphorylation of MeCP2 at Ser421, a site that regulates MeCP2's function as a repressor. Phosphorylation is selectively induced in GABAergic interneurons of the NAc, and its extent strongly predicts the degree of behavioral sensitization. These data reveal new roles for MeCP2 both in mesolimbocortical circuit development and in the regulation of psychostimulant-induced behaviors.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Methyl-CpG-Binding Protein 2/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Corpus Striatum/drug effects , Corpus Striatum/physiology , Dendritic Spines/drug effects , Dendritic Spines/physiology , Drinking Behavior/drug effects , Drinking Behavior/physiology , Genes, Immediate-Early/drug effects , Interneurons/drug effects , Interneurons/physiology , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Phosphorylation/drug effects , Synapses/drug effects , Synapses/physiology , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...