Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Neurosci ; 35(10): 4296-305, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25762676

ABSTRACT

Given that the κ opioid receptor (KOR) system has been implicated in psychostimulant abuse, we evaluated whether the selective KOR antagonist norbinaltorphimine dihydrochloride (nor-BNI) would attenuate the escalation of methamphetamine (METH) intake in an extended-access self-administration model. Systemic nor-BNI decreased the escalation of intake of long-access (LgA) but not short-access (ShA) self-administration. nor-BNI also decreased elevated progressive-ratio (PR) breakpoints in rats in the LgA condition and continued to decrease intake after 17 d of abstinence, demonstrating that the effects of a nor-BNI injection are long lasting. Rats with an ShA history showed an increase in prodynorphin immunoreactivity in both the nucleus accumbens (NAc) core and shell, but LgA animals showed a selective increase in the NAc shell. Other cohorts of rats received nor-BNI directly into the NAc shell or core and entered into ShA or LgA. nor-BNI infusion in the NAc shell, but not NAc core, attenuated escalation of intake and PR responding for METH in LgA rats. These data indicate that the development and/or expression of compulsive-like responding for METH under LgA conditions depends on activation of the KOR system in the NAc shell and suggest that the dynorphin-KOR system is a central component of the neuroplasticity associated with negative reinforcement systems that drive the dark side of addiction.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Methamphetamine/administration & dosage , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Receptors, Opioid, kappa/metabolism , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Enkephalins/metabolism , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Protein Precursors/metabolism , Rats , Rats, Wistar , Reinforcement, Psychology , Self Administration
2.
PLoS One ; 9(5): e97216, 2014.
Article in English | MEDLINE | ID: mdl-24816773

ABSTRACT

Dynorphins, endogenous opioid peptides that arise from the precursor protein prodynorphin (Pdyn), are hypothesized to be involved in the regulation of mood states and the neuroplasticity associated with addiction. The current study tested the hypothesis that dynorphin in the nucleus accumbens (NAcc) mediates such effects. More specifically, we examined whether knockdown of Pdyn within the NAcc in rats would alter the expression of depressive-like and anxiety-like behavior, as well as cocaine locomotor sensitization. Wistar rats were injected with adeno-associated viral (AAV) vectors encoding either a Pdyn-specific short hairpin RNA (AAV-shPdyn) or a scrambled shRNA (AAV-shScr) as control. Four weeks later, rats were tested for anxiety-like behavior in the elevated plus maze test and depressive-like behavior in the forced swim test (FST). Finally, rats received one daily injection of saline or cocaine (20 mg/kg, i.p.), followed by assessment of locomotion for 4 consecutive days. Following 3 days of abstinence, the rats completed 2 additional daily cocaine/saline locomotor trials. Pdyn knockdown in the NAcc led to a significant reduction in depressive-like behavior in the FST, but had no effect on anxiety-like behavior in the elevated plus maze. Pdyn knockdown did not alter baseline locomotor behavior, the locomotor response to acute cocaine, or the initial sensitization of the locomotor response to cocaine over the first 4 cocaine treatment days. However, following 3 days abstinence the locomotor response to the cocaine challenge returned to their original levels in the AAV-shPdyn rats while remaining heightened in the AAV-shScr rats. These results suggest that dynorphin in a very specific area of the nucleus accumbens contributes to depressive-like states and may be involved in neuroadaptations in the NAcc that contribute to the development of cocaine addiction as a persistent and lasting condition.


Subject(s)
Cocaine/pharmacology , Depression/drug therapy , Enkephalins/metabolism , Gene Expression Regulation/drug effects , Locomotion/drug effects , Nucleus Accumbens/metabolism , Protein Precursors/metabolism , RNA, Small Interfering/pharmacology , Analysis of Variance , Animals , Anxiety/drug therapy , Dependovirus , Depression/metabolism , Enkephalins/genetics , Gene Knockdown Techniques , Genetic Vectors/genetics , In Situ Hybridization , Maze Learning , Protein Precursors/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar
3.
Neuropharmacology ; 76 Pt B: 370-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23747571

ABSTRACT

Drug addiction has been conceptualized as a chronically relapsing disorder of compulsive drug seeking and taking that progresses through three stages: binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. Drug addiction impacts multiple motivational mechanisms and can be conceptualized as a disorder that progresses from positive reinforcement (binge/intoxication stage) to negative reinforcement (withdrawal/negative affect stage). The construct of negative reinforcement is defined as drug taking that alleviates a negative emotional state. Our hypothesis is that the negative emotional state that drives such negative reinforcement is derived from dysregulation of key neurochemical elements involved in the brain stress systems within the frontal cortex, ventral striatum, and extended amygdala. Specific neurochemical elements in these structures include not only recruitment of the classic stress axis mediated by corticotropin-releasing factor (CRF) in the extended amygdala as previously hypothesized but also recruitment of dynorphin-κ opioid aversive systems in the ventral striatum and extended amygdala. Additionally, we hypothesized that these brain stress systems may be engaged in the frontal cortex early in the addiction process. Excessive drug taking engages activation of CRF not only in the extended amygdala, accompanied by anxiety-like states, but also in the medial prefrontal cortex, accompanied by deficits in executive function that may facilitate the transition to compulsive-like responding. Excessive activation of the nucleus accumbens via the release of mesocorticolimbic dopamine or activation of opioid receptors has long been hypothesized to subsequently activate the dynorphin-κ opioid system, which in turn can decrease dopaminergic activity in the mesocorticolimbic dopamine system. Blockade of the κ opioid system can also block anxiety-like and reward deficits associated with withdrawal from drugs of abuse and block the development of compulsive-like responding during extended access to drugs of abuse, suggesting another powerful brain stress/anti-reward system that contributes to compulsive drug seeking. Thus, brain stress response systems are hypothesized to be activated by acute excessive drug intake, to be sensitized during repeated withdrawal, to persist into protracted abstinence, and to contribute to the development and persistence of addiction. The recruitment of anti-reward systems provides a powerful neurochemical basis for the negative emotional states that are responsible for the dark side of addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.


Subject(s)
Impulsive Behavior/complications , Reinforcement, Psychology , Substance-Related Disorders/complications , Amygdala/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Drug-Seeking Behavior , Dynorphins/metabolism , Humans , Prefrontal Cortex/metabolism
4.
Addict Biol ; 19(1): 77-86, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23461423

ABSTRACT

Abstinence from cocaine self-administration (SA) is associated with neuroadaptations in the prefrontal cortex (PFC) and nucleus accumbens (NAc) that are implicated in cocaine-induced neuronal plasticity and relapse to drug-seeking. Alterations in cAMP-dependent protein kinase A (PKA) signaling are prominent in medium spiny neurons in the NAc after repeated cocaine exposure but it is unknown whether similar changes occur in the PFC. Because cocaine SA induces disturbances in glutamatergic transmission in the PFC-NAc pathway, we examined whether dysregulation of PKA-mediated molecular targets in PFC-NAc neurons occurs during abstinence and, if so, whether it contributes to cocaine-seeking. We measured the phosphorylation of cAMP response element binding protein (Ser133) and GluA1 (Ser845) in the dorsomedial (dm) PFC and the presynaptic marker, synapsin I (Ser9, Ser62/67, Ser603), in the NAc after 7 days of abstinence from cocaine SA with or without cue-induced cocaine-seeking. We also evaluated whether infusion of the PKA inhibitor, 8-bromo-Rp-cyclic adenosine 3', 5'-monophosphorothioate (Rp-cAMPs), into the dmPFC after abstinence would affect cue-induced cocaine-seeking and PKA-regulated phosphoprotein levels. Seven days of forced abstinence increased the phosphorylation of cAMP response element binding protein and GluA1 in the dmPFC and synapsin I (Ser9) in the NAc. Induction of these phosphoproteins was reversed by a cue-induced relapse test of cocaine-seeking. Bilateral intra-dmPFC Rp-cAMPs rescued abstinence-elevated PKA-mediated phosphoprotein levels in the dmPFC and NAc and suppressed cue-induced relapse. Thus, by inhibiting abstinence-induced PKA molecular targets, relapse reverses abstinence-induced neuroadaptations in the dmPFC that are responsible, in part, for the expression of cue-induced cocaine-seeking.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Drug-Seeking Behavior/physiology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Analysis of Variance , Animals , Blotting, Western , CREB-Binding Protein/metabolism , Cocaine/administration & dosage , Cocaine/pharmacology , Cues , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Dopamine Uptake Inhibitors/administration & dosage , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Male , Neuronal Plasticity/drug effects , Phosphoproteins/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Recurrence , Self Administration , Synapsins/metabolism , Thionucleotides/pharmacology
5.
J Neurosci ; 33(49): 19384-92, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305833

ABSTRACT

The abuse of opioid drugs, both illicit and prescription, is a persistent problem in the United States, accounting for >1.2 million users who require treatment each year. Current treatments rely on suppressing immediate withdrawal symptoms and replacing illicit drug use with long-acting opiate drugs. However, the mechanisms that lead to preventing opiate dependence are still poorly understood. We hypothesized that κ opioid receptor (KOR) activation during chronic opioid intake contributes to negative affective states associated with withdrawal and the motivation to take increasing amounts of heroin. Using a 12 h long-access model of heroin self-administration, rats showed escalation of heroin intake over several weeks. This was prevented by a single high dose (30 mg/kg) of the long-acting KOR antagonist norbinaltorphimine (nor-BNI), paralleled by reduced motivation to respond for heroin on a progressive-ratio schedule of reinforcement, a measure of compulsive-like responding. Systemic nor-BNI also significantly decreased heroin withdrawal-associated anxiety-like behavior. Immunohistochemical analysis showed prodynorphin content increased in the nucleus accumbens core in all heroin-exposed rats, but selectively increased in the nucleus accumbens shell in long-access rats. Local infusion of nor-BNI (4 µg/side) into accumbens core altered the initial intake of heroin but not the rate of escalation, while local injection into accumbens shell selectively suppressed increases in heroin intake over time without altering initial intake. These data suggest that dynorphin activity in the nucleus accumbens mediates the increasing motivation for heroin taking and compulsive-like responding for heroin, suggesting that KOR antagonists may be promising targets for the treatment of opioid addiction.


Subject(s)
Heroin Dependence/drug therapy , Heroin Dependence/psychology , Motivation/drug effects , Narcotic Antagonists/pharmacology , Receptors, Opioid, kappa/antagonists & inhibitors , Animals , Anxiety/psychology , Catheterization , Conditioning, Operant , Enkephalins/metabolism , Immunohistochemistry , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Protein Precursors/metabolism , Rats , Rats, Wistar , Reinforcement Schedule , Self Administration , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/psychology
6.
Biol Psychiatry ; 74(7): 520-8, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23751206

ABSTRACT

BACKGROUND: Studies have demonstrated an enhanced dynorphin/kappa-opioid receptor (KOR) system following repeated cocaine exposure, but few reports have focused on neuroadaptations within the central amygdala (CeA). METHODS: We identified KOR-related physiological changes in the CeA following escalation of cocaine self-administration in rats. We used in vitro slice electrophysiological (intracellular and whole-cell recordings) methods to assess whether differential cocaine access in either 1-hour (short access [ShA]) or 6-hour (long access [LgA]) sessions induced plasticity at CeA gamma-aminobutyric acid (GABA)ergic synapses or altered the sensitivity of these synapses to KOR agonism (U50488) or antagonism (norbinaltorphimine [norBNI]). We then determined the functional effects of CeA KOR blockade in cocaine-related behaviors. RESULTS: Baseline evoked GABAergic transmission was enhanced in the CeA from ShA and LgA rats compared with cocaine-naïve rats. Acute cocaine (1 µmol/L) application significantly decreased GABA release in all groups (naïve, ShA, and LgA rats). Application of U50488 (1 µmol/L) significantly decreased GABAergic transmission in the CeA from naïve rats but increased it in LgA rats. Conversely, norBNI (200 nmol/L) significantly increased GABAergic transmission in the CeA from naïve rats but decreased it in LgA rats. Norbinaltorphimine did not alter the acute cocaine-induced inhibition of GABAergic responses. Finally, CeA microinfusion of norBNI blocked cocaine-induced locomotor sensitization and attenuated the heightened anxiety-like behavior observed during withdrawal from chronic cocaine exposure in the defensive burying paradigm. CONCLUSIONS: Together these data demonstrate that CeA dynorphin/KOR systems are dysregulated following excessive cocaine exposure and suggest KOR antagonism as a viable therapeutic strategy for cocaine addiction.


Subject(s)
Amygdala/physiopathology , Cocaine-Related Disorders/physiopathology , Inhibitory Postsynaptic Potentials , Receptors, Opioid, kappa/physiology , gamma-Aminobutyric Acid/physiology , Animals , Cocaine/administration & dosage , Male , Rats , Rats, Wistar , Self Administration
7.
J Neurosci ; 32(22): 7563-71, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22649234

ABSTRACT

Alcoholism is characterized by a compulsion to seek and ingest alcohol, loss of control over intake, and the emergence of a negative emotional state during abstinence. We hypothesized that sustained activation of neuroendocrine stress systems (e.g., corticosteroid release via the hypothalamic-pituitary-adrenal axis) by alcohol intoxication and withdrawal and consequent alterations in glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) activation drive compulsive alcohol drinking. Our results showed that rats exposed to alcohol vapor to the point of dependence displayed increased alcohol intake, compulsive drinking measured by progressive-ratio responding, and persistent alcohol consumption despite punishment, assessed by adding quinine to the alcohol solution, compared with control rats that were not exposed to alcohol vapor. No group differences were observed in the self-administration of saccharin-sweetened water. Acute alcohol withdrawal was accompanied by downregulated GR mRNA in various stress/reward-related brain regions [i.e., prefrontal cortex, nucleus accumbens (NAc), and bed nucleus of the stria terminalis (BNST)], whereas protracted alcohol abstinence was accompanied by upregulated GR mRNA in the NAc core, ventral BNST, and central nucleus of the amygdala. No significant alterations in MR mRNA levels were found. Chronic GR antagonism with mifepristone (RU38486) prevented the escalation of alcohol intake and compulsive responding induced by chronic, intermittent alcohol vapor exposure. Chronic treatment with mifepristone also blocked escalated alcohol drinking and compulsive responding during protracted abstinence. Thus, the GR system appears to be involved in the development of alcohol dependence and may represent a potential pharmacological target for the treatment of alcoholism.


Subject(s)
Alcohol Drinking/metabolism , Brain/metabolism , Receptors, Glucocorticoid/metabolism , Substance Withdrawal Syndrome/metabolism , Up-Regulation , Alcohol Drinking/drug therapy , Alcohol Drinking/pathology , Analysis of Variance , Animals , Behavior, Addictive/drug therapy , Behavior, Addictive/metabolism , Central Nervous System Depressants/administration & dosage , Compulsive Behavior/physiopathology , Conditioning, Operant/drug effects , Ethanol/administration & dosage , Hormone Antagonists/therapeutic use , Male , Mifepristone/therapeutic use , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Reinforcement Schedule , Self Administration , Substance Withdrawal Syndrome/drug therapy , Up-Regulation/drug effects
8.
J Neurosci ; 31(3): 834-42, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21248106

ABSTRACT

Cocaine-mediated neuroadaptations in the prefrontal cortical-nucleus accumbens pathway underlie drug-seeking in animals with a cocaine self-administration (SA) history. Neuroplasticity in the cortico-accumbens pathway is regulated, in part, by the expression and availability of neurotrophic factors, such as BDNF. We have previously demonstrated that infusion of BDNF into the dorsomedial prefrontal cortex (dmPFC) immediately after the last of 10 cocaine SA sessions attenuates contextual, cue- and cocaine prime-induced reinstatement of cocaine-seeking (Berglind et al., 2007) and normalizes cocaine-induced disruption of glutamatergic transmission in the nucleus accumbens (Berglind et al., 2009). In the present study, the suppressive effect of intra-dmPFC BDNF on cocaine-seeking is shown to depend on Trk receptor-mediated activation of extracellular signal-regulated kinase (ERK) signaling in the dmPFC. The tyrosine kinase inhibitor, K252a, and the mitogen-activated protein/extracellular signal-regulated kinase kinase inhibitor, U0126 (1,4-diamino-2,3-dicyano-1,4-bis[2-aminophenylthio]butadiene), prevented BDNF's suppressive effects on cocaine-seeking. Vehicle-infused rats with a cocaine SA history showed significant decreases in ERK and cyclic AMP response element binding protein (CREB), but not Akt, phosphorylation after the final cocaine SA session that were reversed by intra-dmPFC BDNF. Additionally, BDNF's ability to normalize cocaine-mediated decreases in ERK and CREB phosphorylation was blocked by U0126, demonstrating that ERK/MAPK activation mediated the behavioral effects. This study elucidates a mechanism whereby BDNF/TrkB (tropomyosin receptor kinase B) activates ERK-regulated CREB phosphorylation in the dmPFC to counteract the neuroadaptations induced by cocaine SA and subsequent relapse to cocaine-seeking.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cocaine/administration & dosage , Drug-Seeking Behavior/drug effects , Mitogen-Activated Protein Kinases/metabolism , Prefrontal Cortex/drug effects , Receptor, trkB/metabolism , Analysis of Variance , Animals , Blotting, Western , Cues , Drug-Seeking Behavior/physiology , Male , Motivation/drug effects , Motivation/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Self Administration
9.
Brain Res ; 1314: 183-93, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-19732758

ABSTRACT

The effects of brain-derived neurotrophic factor (BDNF) on cocaine-seeking are brain region-specific. Infusion of BDNF into subcortical structures, like the nucleus accumbens and ventral tegmental area, enhances cocaine-induced behavioral sensitization and cocaine-seeking. Conversely, repeated administration of BDNF antiserum into the nucleus accumbens during chronic cocaine self-administration attenuates cocaine-induced reinstatement. In contrast, BDNF infusion into the dorsomedial prefrontal cortex immediately following a final session of cocaine self-administration attenuates relapse to cocaine-seeking after abstinence, as well as cue- and cocaine prime-induced reinstatement of cocaine-seeking following extinction. BDNF-induced alterations in the ERK-MAP kinase cascade and in prefronto-accumbens glutamatergic transmission are implicated in BDNF's ability to alter cocaine-seeking. Within 22 hours after infusion into the prefrontal cortex, BDNF increases BDNF protein in prefrontal cortical targets, including nucleus accumbens, and restores cocaine-mediated decreases in phospho-ERK expression in the nucleus accumbens. Furthermore, 3 weeks after BDNF infusion in animals with a cocaine self-administration history, suppressed basal levels of glutamate are normalized and a cocaine prime-induced increase in extracellular glutamate levels in the nucleus accumbens is prevented. Thus, BDNF may have local effects at the site of infusion and distal effects in target areas that are critical to mediating or preventing cocaine-induced dysfunctional neuroadaptations.


Subject(s)
Brain/drug effects , Brain/physiopathology , Cocaine-Related Disorders/physiopathology , Cocaine/pharmacology , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/physiology , Cocaine-Related Disorders/metabolism , Dopamine Uptake Inhibitors/pharmacology , Glutamic Acid/metabolism , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiopathology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology
10.
J Neurosci ; 29(12): 3715-9, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19321768

ABSTRACT

The glutamatergic pathway arising in the dorsomedial prefrontal cortex (dmPFC) and projecting to the nucleus accumbens (NAc) core is a critical component of the reward circuitry that underlies reinstatement to cocaine-seeking behavior. Brain-derived neurotrophic factor (BDNF) is expressed by and modulates PFC-NAc neurons. BDNF infusion into the dmPFC attenuates reinstatement to cocaine-seeking behavior, as well as some cocaine-induced molecular adaptations within the NAc. In the present study, it is demonstrated that a single intra-dmPFC infusion of BDNF prevents cocaine self-administration-induced reduction in basal extracellular glutamate, as well as cocaine prime-induced increases in extracellular glutamate levels within the NAc. These data suggest that intra-PFC BDNF attenuates reinstatement to cocaine-seeking behavior by normalizing cocaine-induced neuroadaptations that alter glutamate neurotransmission within the NAc.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Cocaine-Related Disorders/metabolism , Extracellular Space/metabolism , Glutamic Acid/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cocaine/administration & dosage , Conditioning, Operant , Extinction, Psychological , Male , Rats , Rats, Sprague-Dawley , Self Administration , Synaptic Transmission
11.
Eur J Neurosci ; 26(3): 757-66, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17651427

ABSTRACT

The medial prefrontal cortex (mPFC) is critical for reinstatement of cocaine seeking and is the main source of brain-derived neurotrophic factor (BDNF) to striatal regions of the brain relapse circuitry. To test the hypothesis that BDNF in the mPFC regulates cocaine-seeking behavior, rats were trained to press a lever for cocaine infusions (0.2 mg/inf, 2 h/day) paired with light+tone conditioned stimulus (CS) presentations on 10 consecutive days. After the last self-administration session, rats received a single infusion of BDNF (0.75 microg/0.5 microL/side) into the mPFC; this manipulation produced protracted effects on cocaine-seeking behavior (non-reinforced lever pressing). BDNF pretreatment administered after the last session attenuated cocaine seeking 22 h later and, remarkably, it also blocked cocaine-induced suppression of phospho-extracellular-regulated kinase and elevated BDNF immunoreactivity in the nucleus accumbens. The same pretreatment also suppressed cocaine-seeking behavior elicited by response-contingent CS presentations after 6 days of forced abstinence or extinction training, as well as a cocaine challenge injection (10 mg/kg, i.p.) after extinction training. However, BDNF infused into the mPFC had no effect on food-seeking behavior. Furthermore, BDNF infused on the sixth day of abstinence failed to alter responding, suggesting that the regulatory influence of BDNF is time limited. The suppressive effects of BDNF infused into the mPFC on cocaine seeking indicate that BDNF regulates cortical pathways implicated in relapse to drug seeking and that corticostriatal BDNF adaptations during early abstinence diminish compulsive drug seeking.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cocaine-Related Disorders/metabolism , Cocaine/adverse effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/administration & dosage , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/physiopathology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dopamine Uptake Inhibitors/adverse effects , Drug Administration Routes , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/physiopathology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiopathology , Prefrontal Cortex/physiopathology , Rats , Rats, Sprague-Dawley , Secondary Prevention , Self Administration , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...