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










Publication year range
1.
PLoS One ; 11(8): e0160948, 2016.
Article in English | MEDLINE | ID: mdl-27505161

ABSTRACT

RACK1 is a scaffolding protein that contributes to the specificity and propagation of several signaling cascades including the cAMP pathway. As such, RACK1 participates in numerous cellular functions ranging from cell migration and morphology to gene transcription. To obtain further insights on the mechanisms whereby RACK1 regulates cAMP-dependent processes, we set out to identify new binding partners of RACK1 during activation of the cAMP signaling using a proteomics strategy. We identified ß-actin as a direct RACK1 binding partner and found that the association between ß-actin and RACK1 is increased in response to the activation of the cAMP pathway. Furthermore, we show that cAMP-dependent increase in BDNF expression requires filamentous actin. We further report that ß-actin associates with the BDNF promoter IV upon the activation of the cAMP pathway and present data to suggest that the association of ß-actin with BDNF promoter IV is RACK1-dependent. Taken together, our data suggest that ß-actin is a new RACK1 binding partner and that the RACK1 and ß-actin association participate in the cAMP-dependent regulation of BDNF transcription.


Subject(s)
Actins/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Actin Cytoskeleton/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Protein Binding , Protein Transport , Receptors for Activated C Kinase , Transcription, Genetic
2.
J Biol Chem ; 287(1): 322-336, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22069327

ABSTRACT

RACK1 is a scaffolding protein that spatially and temporally regulates numerous signaling cascades. We previously found that activation of the cAMP signaling pathway induces the translocation of RACK1 to the nucleus. We further showed that nuclear RACK1 is required to promote the transcription of the brain-derived neurotrophic factor (BDNF). Here, we set out to elucidate the mechanism underlying cAMP-dependent RACK1 nuclear translocation and BDNF transcription. We identified the scaffolding protein 14-3-3ζ as a direct binding partner of RACK1. Moreover, we found that 14-3-3ζ was necessary for the cAMP-dependent translocation of RACK1 to the nucleus. We further observed that the disruption of RACK1/14-3-3ζ interaction with a peptide derived from the RACK1/14-3-3ζ binding site or shRNA-mediated 14-3-3ζ knockdown inhibited cAMP induction of BDNF transcription. Together, these data reveal that the function of nuclear RACK1 is mediated through its interaction with 14-3-3ζ. As RACK1 and 14-3-3ζ are two multifunctional scaffolding proteins that coordinate a wide variety of signaling events, their interaction is likely to regulate other essential cellular functions.


Subject(s)
14-3-3 Proteins/metabolism , Brain-Derived Neurotrophic Factor/genetics , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Rats , Receptors for Activated C Kinase , Receptors, Cell Surface/chemistry , Reproducibility of Results , Signal Transduction
3.
Addict Biol ; 15(4): 424-33, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21040239

ABSTRACT

Ibogaine is a naturally occurring alkaloid that has been reported to decrease various adverse phenotypes associated with exposure to drugs of abuse and alcohol in human and rodent models. Unfortunately, ibogaine cannot be used as a medication to treat addiction because of severe side effects. Previously, we reported that the desirable actions of ibogaine to reduce self-administration of, and relapse to, alcohol consumption are mediated via the upregulation of the expression of the glial cell line-derived neurotrophic factor (GDNF) in the midbrain ventral tegmental area (VTA), and the consequent activation of the GDNF pathway. The ibogaine metabolite, noribogaine, and a synthetic derivative of ibogaine, 18-Methoxycoronaridine (18-MC), possess a similar anti-addictive profile as ibogaine in rodent models, but without some of its adverse side effects. Here, we determined whether noribogaine and/or 18-MC, like ibogaine, increase GDNF expression, and whether their site of action to reduce alcohol consumption is the VTA. We used SH-SY5Y cells as a cell culture model and found that noribogaine, like ibogaine, but not 18-MC, induces a robust increase in GDNF mRNA levels. Next, we tested the effect of intra-VTA infusion of noribogaine and 18-MC on rat operant alcohol self-administration and found that noribogaine, but not 18-MC, in the VTA decreases responding for alcohol. Together, our results suggest that noribogaine and 18-MC have different mechanisms and sites of action.


Subject(s)
Alcohol Deterrents/pharmacology , Alcohol Drinking/genetics , Alcoholism/genetics , Gene Expression/drug effects , Gene Expression/genetics , Ibogaine/analogs & derivatives , Ibogaine/pharmacology , Ventral Tegmental Area/drug effects , Animals , Cell Line, Tumor , Conditioning, Operant/drug effects , Humans , Male , Motivation/drug effects , RNA, Messenger/genetics , Rats , Rats, Long-Evans , Self Administration
4.
J Neurosci ; 30(43): 14502-12, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20980608

ABSTRACT

Spontaneous firing of ventral tegmental area (VTA) dopamine (DA) neurons provides ambient levels of DA in target areas such as the nucleus accumbens (NAc) and the prefrontal cortex (PFC). Here we report that the glial cell line-derived neurotrophic factor (GDNF), produced in one target region, the NAc, is retrogradely transported by DA neurons to the VTA where the growth factor positively regulates the spontaneous firing activity of both NAc- and PFC-projecting DA neurons in a mechanism that requires the activation of the mitogen-activated protein kinase (MAPK) pathway. We also show that the consequence of GDNF-mediated activation of the MAPK signaling cascade in the VTA is an increase in DA overflow in the NAc. Together, these results demonstrate that NAc-produced GDNF serves as a retrograde enhancer that upregulates the activity of the mesocorticolimbic DA system.


Subject(s)
Cerebral Cortex/physiology , Dopamine/physiology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Limbic System/physiology , Nucleus Accumbens/metabolism , Animals , Blotting, Western , Brain Chemistry , Cloning, Molecular , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Microdialysis , Mitogen-Activated Protein Kinases/physiology , Nucleus Accumbens/physiology , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sympathectomy, Chemical , Ventral Tegmental Area/metabolism
5.
J Biol Chem ; 285(25): 19043-50, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20410295

ABSTRACT

Scaffolding proteins are major contributors to the spatial and temporal orchestration of signaling cascades and hence cellular functions. RACK1 is a scaffolding protein that plays an important role in the regulation of, and cross-talk between, various signaling pathways. Here we report that RACK1 is a mediator of chromatin remodeling, resulting in an exon-specific expression of the brain-derived neurotrophic factor (BDNF) gene. Specifically, we found that following the activation of the cAMP pathway, nuclear RACK1 localizes at the promoter IV region of the BDNF gene by its association with histones H3 and H4, leading to the dissociation of the transcription repressor methyl-CpG-binding protein 2 (MeCP2) from the promoter, resulting in the acetylation of histone H4. These chromatin modifications lead to the activation of the promoter and to the subsequent promoter-controlled transcription of BDNF exon IV. Our findings expand our knowledge regarding the function of scaffolding proteins such as RACK1. Furthermore, this novel mechanism for the regulation of exon-specific expression of the BDNF gene by RACK1 could have implications on the neuronal functions of the growth factor including synaptic plasticity, learning, and memory.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Epigenesis, Genetic , GTP-Binding Proteins/biosynthesis , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , CpG Islands , Histones/chemistry , Humans , Learning , Memory , Methyl-CpG-Binding Protein 2/genetics , Rats , Rats, Sprague-Dawley , Receptors for Activated C Kinase , Signal Transduction
6.
J Neurosci ; 29(43): 13494-502, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19864562

ABSTRACT

We previously found that brain-derived neurotrophic factor (BDNF)-haplodeficient mice exhibit greater ethanol-induced place preference and psychomotor sensitization, and greater ethanol consumption after deprivation, than control mice. We further observed that, in mice, voluntary ethanol intake increases BDNF expression in the dorsal striatum (DS). Here, we determined whether BDNF within the DS regulates ethanol self-administration in Long-Evans rats trained to self-administer a 10% ethanol solution. We observed a greater increase in BDNF expression after ethanol self-administration in the dorsolateral striatum (DLS) than in the dorsomedial striatum (DMS). We further found that downregulation of endogenous BDNF using viral-mediated siRNA in the DLS, but not in the DMS, significantly increased ethanol self-administration. Infusion of exogenous BDNF (0.25 microg/microl/side into the DMS; 0.25 and 0.75 microg/microl/side into the DLS) attenuated responding for ethanol when infused 3 h before the beginning of the self-administration session. Although the decrease in ethanol intake was similar in the DLS and DMS, BDNF infused in the DLS, but not in the DMS, induced an early termination of the drinking episode. Furthermore, the action of BDNF in the DLS was specific for ethanol, as infusion of the neurotrophic factor in the DMS, but not DLS, resulted in a reduction of sucrose intake. Together, these findings demonstrate that the BDNF pathway within the DLS controls the level of ethanol self-administration. Importantly, our results suggest that an endogenous signaling pathway within the same brain region that mediates drug-taking behavior also plays a critical role in gating the level of ethanol intake.


Subject(s)
Alcohol Drinking/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Central Nervous System Depressants/pharmacology , Corpus Striatum/metabolism , Ethanol/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cell Line, Tumor , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/blood , Corpus Striatum/drug effects , Drinking Behavior/drug effects , Drinking Behavior/physiology , Ethanol/administration & dosage , Ethanol/blood , Gene Knockdown Techniques , Humans , Male , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Rats, Long-Evans , Self Administration , Signal Transduction , Sucrose/administration & dosage , Time Factors
7.
Biol Psychiatry ; 66(2): 146-53, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19232578

ABSTRACT

BACKGROUND: Cabergoline is an ergotamine derivative that increases the expression of glial cell line-derived neurotrophic factor (GDNF) in vitro. We recently showed that GDNF in the ventral tegmental area (VTA) reduces the motivation to consume alcohol. We therefore set out to determine whether cabergoline administration decreases alcohol-drinking and -seeking behaviors via GDNF. METHODS: Reverse transcription polymerase chain reaction (RT-PCR) and Enzyme-Linked ImmunoSorbent Assay (ELISA) were used to measure GDNF levels. Western blot analysis was used for phosphorylation experiments. Operant self-administration in rats and a two-bottle choice procedure in mice were used to assess alcohol-drinking behaviors. Instrumental performance tested during extinction was used to measure alcohol-seeking behavior. The [35S]GTPgammaS binding assay was used to assess the expression and function of the dopamine D2 receptor (D2R). RESULTS: We found that treatment of the dopaminergic-like cell line SH-SY5Y with cabergoline and systemic administration of cabergoline in rats resulted in an increase in GDNF level and in the activation of the GDNF pathway. Cabergoline treatment decreased alcohol-drinking and -seeking behaviors including relapse, and its action to reduce alcohol consumption was localized to the VTA. Finally, the increase in GDNF expression and the decrease in alcohol consumption by cabergoline were abolished in GDNF heterozygous knockout mice. CONCLUSIONS: Together, these findings suggest that cabergoline-mediated upregulation of the GDNF pathway attenuates alcohol-drinking behaviors and relapse. Alcohol abuse and addiction are devastating and costly problems worldwide. This study puts forward the possibility that cabergoline might be an effective treatment for these disorders.


Subject(s)
Alcohol Deterrents , Alcohol Drinking/drug therapy , Alcohol Drinking/psychology , Dopamine Agonists/pharmacology , Ergolines/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Animals , Cabergoline , Conditioning, Operant/drug effects , Dopamine Agonists/administration & dosage , Enzyme-Linked Immunosorbent Assay , Ergolines/administration & dosage , Extinction, Psychological/drug effects , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Injections, Intraperitoneal , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Knockout , Microinjections , Rats , Self Administration , Substantia Nigra/physiology
8.
J Neurosci ; 29(2): 543-9, 2009 Jan 14.
Article in English | MEDLINE | ID: mdl-19144854

ABSTRACT

Alcohol has subjective and behavioral effects at the pharmacological levels typically reached during the consumption of one or two alcoholic drinks. Here we provide evidence that an alpha4-subunit-containing GABA(A) receptor contributes to the consumption of low-to-moderate levels of alcohol. Using viral-mediated RNA interference (RNAi), we found that reduced expression of the alpha4 subunit in the nucleus accumbens (NAc) shell of rats decreased their free consumption of and preference for alcohol. The time course for the reduced alcohol intake paralleled the time course of alpha4 mRNA reductions achieved after viral-mediated RNAi for alpha4. Furthermore, the reduction in drinking was region- and alcohol-specific: there was no effect of reductions in alpha4 expression in the NAc core on alcohol intake, and reductions in alpha4 expression in the NAc shell did not alter sucrose or water intake. These results indicate that the GABA(A) receptor alpha4 subunit in the NAc shell mediates alcohol intake.


Subject(s)
Ethanol/administration & dosage , Gene Expression Regulation/drug effects , Nucleus Accumbens/physiology , Receptors, GABA-A/metabolism , Analysis of Variance , Animals , Area Under Curve , Cell Line, Transformed , Chromatography, Gas/methods , Conditioning, Operant/physiology , Eating/genetics , Eating/physiology , Ethanol/blood , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Humans , Male , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Long-Evans , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Time Factors , Transfection/methods
9.
J Biol Chem ; 283(19): 12811-8, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18343820

ABSTRACT

We previously found that glial cell line-derived neurotrophic factor (GDNF) in the midbrain ventral tegmental area (VTA) negatively regulates alcohol drinking (He, D. Y., McGough, N. N., Ravindranathan, A., Jeanblanc, J., Logrip, M. L., Phamluong, K., Janak, P. H., and Ron, D. (2005) J. Neurosci. 25, 619-628). Several studies suggest a role for GDNF in the regulation of tyrosine hydroxylase (TH) levels in the midbrain (Georgievska, B., Kirik, D., and Bjorklund, A. (2004) J. Neurosci. 24, 6437-6445). Up-regulation of TH levels has been reported as a hallmark of biochemical adaptations to in vivo chronic exposure to drugs of abuse, including ethanol (Ortiz, J., Fitzgerald, L. W., Charlton, M., Lane, S., Trevisan, L., Guitart, X., Shoemaker, W., Duman, R. S., and Nestler, E. J. (1995) Synapse 21, 289-298). We hypothesized that GDNF plays an important role in regulating prolonged ethanol-mediated increases in TH protein levels. Using the SH-SY5Y dopaminergic-like cell line, we found that the increase in TH levels in the presence of ethanol required the activation of the cAMP/PKA pathway and was reversed by GDNF. Ethanol treatment did not alter the mRNA level or protein translation of TH, but enhanced the stability of the protein that was decreased by GDNF. Interestingly, we observed that ethanol treatment resulted in an increase in TH association with the chaperone heat shock protein (HSP90) that was mediated by the cAMP/PKA pathway and inhibited by GDNF. Taken together, these data suggest that prolonged ethanol exposure leads to increased association of TH and HSP90 via the cAMP/PKA pathway, resulting in the stabilization and subsequent accumulation of TH. GDNF reverses this ethanol-mediated adaptation by inhibiting the interaction of TH with HSP90.


Subject(s)
Ethanol/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , HSP90 Heat-Shock Proteins/metabolism , Tyrosine 3-Monooxygenase/immunology , Tyrosine 3-Monooxygenase/metabolism , Benzoquinones/pharmacology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Protein Binding , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tyrosine 3-Monooxygenase/genetics , Up-Regulation
10.
FASEB J ; 20(13): 2420-2, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17023388

ABSTRACT

We recently showed that the up-regulation of the glial cell line-derived neurotrophic factor (GDNF) pathway in the midbrain, is the molecular mechanism by which the putative anti-addiction drug Ibogaine mediates its desirable action of reducing alcohol consumption. Human reports and studies in rodents have shown that a single administration of Ibogaine results in a long-lasting reduction of drug craving (humans) and drug and alcohol intake (rodents). Here we determine whether, and how, Ibogaine exerts its long-lasting actions on GDNF expression and signaling. Using the dopaminergic-like SHSY5Y cell line as a culture model, we observed that short-term Ibogaine exposure results in a sustained increase in GDNF expression that is mediated via the induction of a long-lasting autoregulatory cycle by which GDNF positively regulates its own expression. We show that the initial exposure of cells to Ibogaine or GDNF results in an increase in GDNF mRNA, leading to protein expression and to the corresponding activation of the GDNF signaling pathway. This, in turn, leads to a further increase in the mRNA level of the growth factor. The identification of a GDNF-mediated, autoregulatory long-lasting feedback loop could have important implications for GDNF's potential value as a treatment for addiction and neurodegenerative diseases.


Subject(s)
Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Ibogaine/pharmacology , Neuroglia/physiology , Cell Line, Tumor , Excitatory Amino Acid Antagonists/pharmacology , Hallucinogens/pharmacology , Homeostasis/drug effects , Humans , Neuroblastoma , Neuroglia/drug effects
11.
J Neurosci ; 26(5): 1457-64, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452669

ABSTRACT

We recently identified a homeostatic pathway that inhibits ethanol intake. This protective pathway consists of the scaffolding protein RACK1 and brain-derived neurotrophic factor (BDNF). RACK1 translocates to the nucleus after exposure of neurons to ethanol and increases expression of BDNF (McGough et al., 2004). We also found that increasing the levels of BDNF via systemic administration of RACK1 expressed as a Tat-fusion protein (Tat-RACK1) reduces ethanol consumption, whereas reduction of BDNF levels augments this behavior (McGough et al., 2004). Based on these results, we hypothesized that activation of the BDNF receptor TrkB is necessary for the effects of BDNF on ethanol intake and that gene products downstream of BDNF negatively regulate ethanol consumption. Here, we show that inhibition of the BDNF receptor TrkB increases voluntary ethanol consumption in wild-type mice but not in mice lacking one copy of the BDNF gene (BDNF(+/-)). We also find that increases in the levels of BDNF, mediated by ethanol or RACK1, lead to increased dorsal striatal levels of the dopamine D3 receptor (D3R), a gene downstream of BDNF, via activation of the TrkB receptor. Finally, we show that the Tat-RACK1-mediated reduction of ethanol consumption is attenuated by coinjection with either the Trk inhibitor K252a or the dopamine D3R-prefering antagonist U-99194A [5, 6-dimethoxy-2-(di-n-propylamino)indan], suggesting that activation of the BDNF pathway via RACK1 leads to increased expression of the dopamine D3R, which in turn mediates the attenuation of ethanol consumption.


Subject(s)
Alcohol Drinking/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Ethanol/administration & dosage , Receptors, Cell Surface/metabolism , Receptors, Dopamine D3/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Female , Homeostasis , Injections , Male , Mice , Mice, Knockout , Neostriatum/drug effects , Neostriatum/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Receptors for Activated C Kinase , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , Signal Transduction
12.
Neuropsychopharmacology ; 31(4): 768-77, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16160706

ABSTRACT

Homer proteins modulate neuroplasticity in excitatory synapses and are dynamically regulated by cocaine. Whereas acute cocaine elevates immediate-early gene (short) isoforms of Homer1 in the nucleus accumbens, withdrawal from repeated cocaine administration downregulates the expression of constitutive Homer1 isoforms. The present study determined whether or not this downregulation in constitutive Homer expression in the accumbens is necessary for enduring alterations in cocaine-induced changes in the brain and behavior. The long vs short Homer isoforms were overexpressed in the rat nucleus accumbens during drug abstinence, and the adaptations elicited by repeated cocaine on glutamate transmission and motor behavior were measured. It was found that both chronic and acute overexpression of constitutive, but not short, Homer isoforms abolished cocaine-induced sensitization of locomotor hyperactivity and prevented the development of glutamate abnormalities in the accumbens, including the reduction in basal extracellular glutamate content and the sensitized glutamate response to a subsequent cocaine challenge injection. Together, these data indicate that the enduring reduction of long Homer isoforms in the nucleus accumbens of cocaine-withdrawn rats is necessary for the expression of cocaine-induced neuroplasticity.


Subject(s)
Anesthetics, Local/administration & dosage , Carrier Proteins/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Gene Expression Regulation/physiology , Neuronal Plasticity/drug effects , Animals , Behavior, Animal , Carrier Proteins/classification , Chromatography, High Pressure Liquid/methods , Cocaine-Related Disorders/physiopathology , Dopamine/metabolism , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Homer Scaffolding Proteins , Male , Mice , Microdialysis/methods , Motor Activity/drug effects , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Time Factors
13.
J Neurosci ; 25(3): 619-28, 2005 Jan 19.
Article in English | MEDLINE | ID: mdl-15659598

ABSTRACT

Alcohol addiction manifests as uncontrolled drinking despite negative consequences. Few medications are available to treat the disorder. Anecdotal reports suggest that ibogaine, a natural alkaloid, reverses behaviors associated with addiction including alcoholism; however, because of side effects, ibogaine is not used clinically. In this study, we first characterized the actions of ibogaine on ethanol self-administration in rodents. Ibogaine decreased ethanol intake by rats in two-bottle choice and operant self-administration paradigms. Ibogaine also reduced operant self-administration of ethanol in a relapse model. Next, we identified a molecular mechanism that mediates the desirable activities of ibogaine on ethanol intake. Microinjection of ibogaine into the ventral tegmental area (VTA), but not the substantia nigra, reduced self-administration of ethanol, and systemic administration of ibogaine increased the expression of glial cell line-derived neurotrophic factor (GDNF) in a midbrain region that includes the VTA. In dopaminergic neuron-like SHSY5Y cells, ibogaine treatment upregulated the GDNF pathway as indicated by increases in phosphorylation of the GDNF receptor, Ret, and the downstream kinase, ERK1 (extracellular signal-regulated kinase 1). Finally, the ibogaine-mediated decrease in ethanol self-administration was mimicked by intra-VTA microinjection of GDNF and was reduced by intra-VTA delivery of anti-GDNF neutralizing antibodies. Together, these results suggest that GDNF in the VTA mediates the action of ibogaine on ethanol consumption. These findings highlight the importance of GDNF as a new target for drug development for alcoholism that may mimic the effect of ibogaine against alcohol consumption but avoid the negative side effects.


Subject(s)
Alcoholism/physiopathology , Ethanol/pharmacology , Ibogaine/pharmacology , Mesencephalon/drug effects , Nerve Growth Factors/physiology , Alcoholism/drug therapy , Animals , Cell Line, Tumor , Cell Survival/drug effects , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dopamine/physiology , Ethanol/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor , Humans , Ibogaine/administration & dosage , Male , Mesencephalon/metabolism , Mesencephalon/physiology , Mice , Mice, Inbred C57BL , Microinjections , Nerve Growth Factors/biosynthesis , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Recurrence , Self Administration , Substantia Nigra/drug effects , Ventral Tegmental Area/drug effects
14.
J Neurosci ; 24(46): 10542-52, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548669

ABSTRACT

Alcoholism is a devastating disease that manifests as uncontrolled drinking. Consumption of alcohol is regulated by neurochemical systems within specific neural circuits, but endogenous systems that may counteract and thus suppress the behavioral effects of ethanol intake are unknown. Here we demonstrate that BDNF plays a role in reducing the behavioral effects of ethanol, including consumption, in rodents. We found that decreasing the levels of BDNF leads to increased behavioral responses to ethanol, whereas increases in the levels of BDNF, mediated by the scaffolding protein RACK1, attenuate these behaviors. Interestingly, we found that acute exposure of neurons to ethanol leads to increased levels of BDNF mRNA via RACK1. Importantly, acute systemic administration of ethanol and voluntary ethanol consumption lead to increased levels of BDNF expression in the dorsal striatum. Taken together, these findings suggest that RACK1 and BDNF are part of a regulatory pathway that opposes adaptations that lead to the development of alcohol addiction.


Subject(s)
Alcohol Drinking/psychology , Brain-Derived Neurotrophic Factor/physiology , Receptors, Cell Surface/physiology , Alcohol Drinking/metabolism , Alcoholism/metabolism , Alcoholism/psychology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/biosynthesis , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Ethanol/administration & dosage , Ethanol/pharmacology , Gene Products, tat/genetics , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Homeostasis , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/pharmacology , Self Administration
15.
Alcohol Clin Exp Res ; 27(10): 1599-605, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14574230

ABSTRACT

BACKGROUND: Previously, we found that acute ethanol induces the translocation of the scaffolding protein RACK1 to the nucleus. Recently, we found that nuclear RACK1 mediates acute ethanol induction of immediate early gene c-fos expression. Alterations in gene expression are thought to lead to long-term changes that ultimately contribute to the development of alcohol addiction and toxicity. Therefore, we sought to determine the effects of chronic exposure of cells to ethanol on the cellular compartmentalization of RACK1 and on c-fos messenger RNA (mRNA) and protein expression. METHODS: Rat C6 glioma cells were used as the cell culture model. Immunohistochemistry was implemented to visualize the localization of RACK1 and to monitor the protein level of c-fos. Reverse-transcription polymerase chain reaction was used to measure c-fos mRNA levels. The Tat-protein transduction method was used to transduce recombinant Tat-RACK1 into cells as previously described. RESULTS: Chronic exposure of cells to 200 mM ethanol for 24 and 48 hr resulted in the gradual re-distribution of RACK1 out of the nucleus. It is interesting to note that acute ethanol re-challenge immediately after chronic treatment did not result in RACK1 translocation to the nucleus, and nuclear compartmentalization of RACK1 in response to acute ethanol was detected only after 24 hr of withdrawal. Similar patterns were obtained for c-fos expression. Chronic exposure to ethanol did not result in an increase in mRNA or protein levels of c-fos. Furthermore, acute ethanol exposure did not increase c-fos protein levels in cells that were first treated chronically with ethanol. However, transduction of exogenous RACK1 expressed as a Tat-fusion protein was able to rescue c-fos mRNA expression after chronic ethanol exposure. CONCLUSIONS: Our data suggest that RACK1 nuclear compartmentalization and ethanol-induced c-fos expression are transient and are desensitized to ethanol during prolonged exposure to high concentrations. The desensitization is temporary, and RACK1 can respond to acute ethanol treatment after a 24-hr withdrawal period. Our data further suggest that the altered compartmentalization of RACK1 leads to differences in c-fos expression upon acute or chronic exposure to ethanol. In summary, RACK1 is an important molecular mediator of the acute and chronic actions of ethanol on the expression of c-fos. These findings could have implications for the molecular signaling pathways leading to pathologic states associated with alcoholism, including toxicity.


Subject(s)
Adaptation, Physiological/drug effects , Cell Compartmentation/drug effects , Ethanol/administration & dosage , Neoplasm Proteins/physiology , Adaptation, Physiological/physiology , Animals , Cell Compartmentation/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , GTP-Binding Proteins , Genes, fos/drug effects , Genes, fos/physiology , Rats , Receptors for Activated C Kinase , Receptors, Cell Surface
16.
J Biol Chem ; 278(11): 9630-8, 2003 Mar 14.
Article in English | MEDLINE | ID: mdl-12524444

ABSTRACT

We recently identified a novel mechanism for modulation of the phosphorylation state and function of the N-methyl-d-aspartate (NMDA) receptor via the scaffolding protein RACK1. We found that RACK1 binds both the NR2B subunit of the NMDA receptor and the nonreceptor protein-tyrosine kinase, Fyn. RACK1 inhibits Fyn phosphorylation of NR2B and decreases NMDA receptor-mediated currents in CA1 hippocampal slices (Yaka, R., Thornton, C., Vagts, A. J., Phamluong, K., Bonci, A., and Ron, D. (2002) Proc. Natl. Acad. Sci. U. S. A. 99, 5710-5715). Here, we identified the signaling cascade by which RACK1 is released from the NMDA receptor complex and identified the consequences of the dissociation. We found that activation of the cAMP/protein kinase A pathway in hippocampal slices induced the release of RACK1 from NR2B and Fyn. This resulted in the induction of NR2B phosphorylation and the enhancement of NMDA receptor-mediated activity via Fyn. We identified the neuropeptide, pituitary adenylate cyclase activating polypeptide (PACAP(1-38)), as a ligand that induced phosphorylation of NR2B and enhanced NMDA receptor potentials. Finally, we found that activation of the cAMP/protein kinase A pathway induced the movement of RACK1 to the nuclear compartment in dissociated hippocampal neurons. Nuclear RACK1 in turn was found to regulate the expression of brain-derived neurotrophic factor induced by PACAP(1-38). Taken together our results suggest that activation of adenylate cyclase by PACAP(1-38) results in the release of RACK1 from the NMDA receptor and Fyn. This in turn leads to NMDA receptor phosphorylation, enhanced activity mediated by Fyn, and to the induction of brain-derived neurotrophic factor expression by RACK1.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , N-Methylaspartate/chemistry , Neuropeptides/chemistry , Peptides/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Nucleus/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Electrophysiology , Enzyme Activation , Hippocampus/cytology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Models, Biological , N-Methylaspartate/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Phosphorylation , Pituitary Adenylate Cyclase-Activating Polypeptide , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , Rats , Rats, Sprague-Dawley , Receptors for Activated C Kinase , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Tyrosine/metabolism
17.
Mol Pharmacol ; 62(2): 272-80, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12130678

ABSTRACT

Scaffolding proteins such as receptor for activated C kinase (RACK) 1 are involved in the targeting of signaling proteins and play an important role in the regulation of signal transduction cascades. Recently, we found that in cultured cells and in vivo, acute ethanol exposure induces the nuclear compartmentalization of RACK1. To elucidate a physiological role for nuclear RACK1, the Tat protein transduction system was used to transduce RACK1 and RACK1-derived fragments into C6 glioma cells. We found that nuclear RACK1 is mediating the induction of the immediate early gene c-fos expression induced by ethanol. First, transduction of full-length RACK1 (Tat-RACK1) resulted in the induction of c-fos expression and enhancement of ethanol activities. Second, we determined that the C terminus of RACK1 (Tat-RACK1DeltaN) is mediating transcription. Third, we identified a dominant negative fragment of RACK1 that inhibited the nuclear compartmentalization of endogenous RACK1 and inhibited ethanol-induction of c-fos mRNA and protein expression. Last, acute exposure to ethanol or transduction of full-length Tat-RACK1 resulted in an increase in mRNA levels of an activator protein 1 site-containing gene, PAC1 (pituitary adenylate cyclase-activating polypeptide receptor type I), suggesting that nuclear RACK1 is involved in the regulation of the expression of genes that are altered upon acute ethanol treatment. These results may therefore have important implications for the study of alcohol addiction.


Subject(s)
Cell Compartmentation/drug effects , Ethanol/pharmacology , Gene Expression/drug effects , Peptides/metabolism , Animals , Cell Nucleus , Neuropeptides/genetics , Neuropeptides/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Receptors for Activated C Kinase , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...