Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Psychedelic Med (New Rochelle) ; 1(2): 111-119, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37360328

ABSTRACT

Background: The potential use of psychedelic drugs as therapeutics for neuropsychiatric disorders has been limited by their hallucinogenic properties. To overcome this limitation, we developed and characterized tabernanthalog (TBG), a novel analogue of the indole alkaloids ibogaine and 5-methoxy-N,N-dimethyltryptamine with reduced cardiac arrhythmogenic risk and a lack of classical psychedelic drugs-induced sensory alterations. We previously demonstrated that TBG has therapeutic efficacy in a preclinical model of opioid use disorder (OUD) in rats and in a binge model of alcohol drinking in mice. Alcohol is commonly co-used in ∼35-50% of individuals with OUD, and yet, preclinical models that recapitulate this comorbidity are lacking. Methodology: Here we employed a polydrug model of heroin and alcohol couse to screen the therapeutic efficacy of TBG on metrics of both opioid and alcohol seeking. We first exposed rats to alcohol (or control sucrose-fade solution) in the home-cage (HC), using a two-bottle binge protocol, over a period of 1 month. Rats were then split into two groups that underwent self-administration training for either intravenous heroin or oral alcohol, so that we could assess the impact of HC alcohol exposure on the self-administration of each substance separately. Thereafter, rats began self-administering both heroin and alcohol in the same sessions. Finally, we tested the effects of TBG on break points for heroin and alcohol in a progressive ratio test, where the number of lever presses required to obtain a single reward increased exponentially. Results and Conclusion: TBG effectively reduced motivation for heroin and alcohol in this test, indicating its efficacy is preserved in animals with a history of heroin and alcohol polydrug use.

2.
Transl Psychiatry ; 12(1): 432, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36195606

ABSTRACT

As opioid-related fatalities continue to rise, the need for novel opioid use disorder (OUD) treatments could not be more urgent. Two separate hypothalamic neuropeptide systems have shown promise in preclinical OUD models. The oxytocin system, originating in the paraventricular nucleus (PVN), may protect against OUD severity. By contrast, the orexin system, originating in the lateral hypothalamus (LH), may exacerbate OUD severity. Thus, activating the oxytocin system or inhibiting the orexin system are potential therapeutic strategies. The specific role of these systems with regard to specific OUD outcomes, however, is not fully understood. Here, we probed the therapeutic efficacy of pharmacological interventions targeting the orexin or oxytocin system on two distinct metrics of OUD severity in rats-heroin choice (versus choice for natural reward, i.e., food) and cued reward seeking. Using a preclinical model that generates approximately equal choice between heroin and food reward, we examined the impact of exogenously administered oxytocin, an oxytocin receptor antagonist (L-368,899), and a dual orexin receptor antagonist (DORA-12) on opioid choice. Whereas these agents did not alter heroin choice when rewards (heroin and food) were available, oxytocin and DORA-12 each significantly reduced heroin seeking in the presence of competing reward cues when no rewards were available. In addition, the number of LH orexin neurons and PVN oxytocin neurons correlated with specific behavioral economic variables indicative of heroin versus food motivation. These data identify a novel bidirectional role of the oxytocin and orexin systems in the ability of opioid-related cues to bias reward seeking.


Subject(s)
Neuropeptides , Oxytocin , Analgesics, Opioid , Animals , Cues , Heroin , Intracellular Signaling Peptides and Proteins , Orexin Receptor Antagonists/pharmacology , Orexins , Oxytocin/pharmacology , Rats , Receptors, Oxytocin
3.
Cell Rep ; 36(8): 109605, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433067

ABSTRACT

Here, we use optogenetics and chemogenetics to investigate the contribution of the paraventricular thalamus (PVT) to nucleus accumbens (NAc) pathway in aversion and heroin relapse in two different heroin self-administration models in rats. In one model, rats undergo forced abstinence in the home cage prior to relapse testing, and in the other, they undergo extinction training, a procedure that is likened to cognitive behavioral therapy. We find that the PVT→NAc pathway is both sufficient and necessary to drive aversion and heroin seeking after abstinence, but not extinction. The ability of extinction to reduce this pathway's contribution to heroin relapse is accompanied by a loss of synaptic plasticity in PVT inputs onto a specific subset of NAc neurons. Thus, extinction may exert therapeutic reductions in opioid seeking by altering synaptic plasticity within the PVT→NAc pathway, resulting in reduced aversion during opioid withdrawal as well as reduced relapse propensity.


Subject(s)
Extinction, Psychological/physiology , Heroin/metabolism , Neuronal Plasticity/physiology , Thalamus/physiology , Animals , Mice , Neurons/metabolism , Nucleus Accumbens/physiology , Rats , Recurrence , Self Administration/methods
4.
Nat Commun ; 12(1): 4788, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373454

ABSTRACT

Activity in numerous brain regions drives heroin seeking, but no circuits that limit heroin seeking have been identified. Furthermore, the neural circuits controlling opioid choice are unknown. In this study, we examined the role of the infralimbic cortex (IL) to nucleus accumbens shell (NAshell) pathway during heroin choice and relapse. This model yielded subpopulations of heroin versus food preferring rats during choice, and choice was unrelated to subsequent relapse rates to heroin versus food cues, suggesting that choice and relapse are distinct behavioral constructs. Supporting this, inactivation of the IL with muscimol produced differential effects on opioid choice versus relapse. A pathway-specific chemogenetic approach revealed, however, that the IL-NAshell pathway acts as a common limiter of opioid choice and relapse. Furthermore, dendritic spines in IL-NAshell neurons encode distinct aspects of heroin versus food reinforcement. Thus, opioid choice and relapse share a common addiction-limiting circuit in the IL-NAshell pathway.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Addictive , Drug-Seeking Behavior/drug effects , Opioid-Related Disorders , Animals , Behavior, Animal , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cues , Decision Making/drug effects , Eating/psychology , Extinction, Psychological/physiology , Food , Heroin/pharmacology , Heroin Dependence , Male , Neural Pathways/physiology , Nucleus Accumbens/metabolism , Rats , Recurrence , Reinforcement, Psychology , Rodentia , Self Administration
5.
Addict Biol ; 26(5): e13012, 2021 09.
Article in English | MEDLINE | ID: mdl-33511707

ABSTRACT

Previous studies have shown that adolescent exposure to cocaine increases drug use in adulthood, albeit incubation of cocaine seeking was found to be attenuated in rats trained to self-administer cocaine during adolescence. We here hypothesize that adolescent exposure to cocaine could alter the rewarding properties of the psychostimulant in adulthood. By employing two of the most widely used animal-experimental-preclinical models to investigate drug addiction, we evaluated whether contingent versus non-contingent cocaine self-administration during adolescence modulates its rewarding threshold in adulthood evaluated by conditioned place preference (CPP). Cocaine self-administration during adolescence increases the rewarding threshold in adulthood; CPP for cocaine was observed at the higher (20 mg/kg), but not at the lower (10 mg/kg), dose employed. Rats exposed to either contingent or non-contingent cocaine during adolescence exhibited the same behavior in the CPP paradigm suggesting that, under our experimental conditions, cocaine rewarding properties are shaped by the psychostimulant itself and not by its motivational effects. From a mechanistic standpoint, the preference for the 20 mg/kg cocaine-paired side in a CPP paradigm appears to depend, at least partially, upon the formation of GluA2-lacking Ca2+ -permeable AMPA receptors and the consequent increase of αCaMKII activity in the NAc, both of which are instead reduced when the 10 mg/kg dose was used. In conclusion, contingent or non-contingent cocaine exposure during adolescence desensitizes adult animals to a rewarding dose of cocaine (10 mg/kg) elevating the rewarding threshold necessary (20 mg/kg) to drive conditioned place preference, an effect that may predispose to higher consumption of cocaine during adulthood.


Subject(s)
Cocaine/pharmacology , Conditioning, Classical/drug effects , Animals , Central Nervous System Stimulants/pharmacology , Female , Male , Motivation , Rats , Receptors, AMPA , Reward , Self Administration
6.
Addict Biol ; 25(4): e12771, 2020 07.
Article in English | MEDLINE | ID: mdl-31132808

ABSTRACT

Nicotine-associated cues can trigger reinstatement in humans as well as in animal models of drug addiction. To date, no behavioral intervention or pharmacological treatment has been effective in preventing relapse in the long term. A large body of evidence indicates that N-acetylcysteine (N-AC) blunts the activation of glutamatergic (GLUergic) neurons in the nucleus accumbens (Nacc) associated with reinstatement. We evaluated the effect of an experimental cue exposure therapy (eCET) alone or in combination with N-AC to verify whether restoring GLU homeostasis enhances extinction of nicotine-associated cues. Rats were trained to associate discriminative stimuli with intravenous nicotine or saline self-administration. Reinforced response was followed by cue signals. After rats met the self-administration criteria, the lasting anti-relapse activity of i.p. N-AC or vehicle was assessed in three different experimental conditions after 14 days of treatment: treatment + eCET; treatment + lever-presses extinction (LP-EXT); and treatment + abstinence. N-AC 100 mg/kg, but not 60 mg/kg, induced anti-relapse activity that persisted 50 days after treatment only when paired with either LP-EXT or eCET with the greater activity found in the latter condition. To identify potential mechanisms for behavioral results, separate groups of rats that received either N-AC or vehicle + eCET were killed at different time points for Nacc Western-blot analysis. Seven days after treatment, chronic N-AC restored the expression of proteins crucial for GLU homeostasis, while at 50 days, it increased the expression of type II metabotropic GLU receptors. These results suggest that N-AC treatment in combination with eCET may offer a novel strategy to prevent relapse in nicotine addiction.


Subject(s)
Acetylcysteine/pharmacology , Cues , Drug-Seeking Behavior/drug effects , Free Radical Scavengers/pharmacology , Glutamic Acid/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Tobacco Use Disorder , Animals , Behavior, Animal , Extinction, Psychological , Implosive Therapy , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Recurrence
7.
Sci Rep ; 9(1): 10709, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31341176

ABSTRACT

The prefrontal cortex is an important regulator of fear expression in humans and rodents. Specifically, the rodent prelimbic (PL) prefrontal cortex drives fear expression during both encoding and retrieval of fear memory. Neuronal ensembles have been proposed to function as memory encoding units, and their re-activation is thought to be necessary for memory retrieval and expression of conditioned behavior. However, it remains unclear whether PL cortex neuronal ensembles that encode fear memory contribute to long-term fear expression during memory retrieval. To address this, we employed a viral-mediated TRAP (Targeted Recombination in Active Population) technology to target PL cortex ensembles active during fear conditioning and expressed the inhibitory Gi-DREADD in fear-encoding ensembles. Male and female rats were trained to lever press for food and subjected to Pavlovian delay fear conditioning, then 28 days later, they underwent a fear memory retrieval test. Chemogenetic inhibition of TRAPed PL cortex ensembles reduced conditioned suppression of food seeking in females, but not males. Neither context nor tone freezing behavior was altered by this manipulation during the same retrieval test. Thus, fear-encoding ensembles in PL cortex drive long-term fear expression in a sex and fear modality dependent manner.


Subject(s)
Fear , Memory, Long-Term , Prefrontal Cortex/physiology , Animals , Conditioning, Classical , Female , Male , Neurons/physiology , Prefrontal Cortex/cytology , Rats , Rats, Wistar
8.
J Psychopharmacol ; 33(3): 372-382, 2019 03.
Article in English | MEDLINE | ID: mdl-30644313

ABSTRACT

BACKGROUND: Environmental conditions have an important function in substance use disorder, increasing or decreasing the risks of relapse. Several studies strongly support the role of the dopamine D2-like and metabotropic glutamate type 5 receptors in maladaptive neurobiological responses to cocaine reward and relapse. AIMS: The present study employed cocaine self-administration with yoked-triad procedure in rats to explore whether drug abstinence in different housing conditions affects the drug-seeking behaviour and the dopamine D2-like and metabotropic glutamate type 5 receptor density and affinity in several regions of the animal brain. METHODS: Rats were trained to self-administer cocaine and later they were forced to abstain either in: (a) enriched environment or (b) isolation cage conditions to evaluate the effect of housing conditions on the drug-seeking behaviour and to assess changes concerning receptors in animals brain. RESULTS: Our results show that exposure to enriched environment conditions strongly reduced active lever presses during cue-induced drug-seeking. At the neurochemical level, we demonstrated a significant increase in the dopamine D2-like receptor density in the prefrontal cortex in animals following drug abstinence in isolation cage or enriched environment conditions, and the reduction in their density in the dorsal striatum provoked by isolation cage conditions. The metabotropic glutamate type 5 receptor density decreased only in the prefrontal cortex after isolation cage and enriched environment abstinence. CONCLUSIONS: This study shows the different impacts caused by the type of housing conditions during abstinence from cocaine self-administration on drug-seeking behaviour in rats. The observed changes in the dopamine D2-like and metabotropic glutamate type 5 receptor Bmax and/or Kd values were brain-region specific and related to either pharmacological and/or motivational features of cocaine.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Dopamine D2/metabolism , Animals , Brain/metabolism , Drug-Seeking Behavior/physiology , Housing, Animal , Male , Rats , Rats, Wistar , Recurrence , Reward , Self Administration
9.
World J Biol Psychiatry ; 20(7): 531-544, 2019 09.
Article in English | MEDLINE | ID: mdl-29380665

ABSTRACT

Objectives: Long-term abstinence following cocaine exposure up-regulates brain-derived neurotrophic factor (BDNF) expression in the mesocorticolimbic pathway. Given the increased vulnerability to drug abuse typical of adolescence, we hypothesized that changes in BDNF expression may become manifest early after the end of cocaine treatment in the adolescent brain.Methods: Rats received cocaine injections from postnatal day 28 (PND28) to PND42 and the mesocorticolimbic expression of BDNF was measured by real-time PCR and Western blotting at PND43.Results: In the ventral tegmental area, BDNF-tropomyosin receptor kinase B (TrΚB) expression and phosphorylation are enhanced while the intracellular signaling is unaltered. In the nucleus accumbens (NAc) shell and core, BDNF and its signaling were down-regulated. In the prelimbic (PL) cortex, we found reduced BDNF expression and increased phosphoprylation of TrΚB, ERK and AKT. In the infralimbic (IL) cortex, increased BDNF expression was coupled with reduced activity and expression of its downstream targets. To evaluate the role of glutamate on BDNF-independent changes, we investigated the expression of the transporter GLT-1 and the activation of the NMDA receptor subunit GluN2B, which were both increased in the PL cortex while reduced in the IL cortex.Conclusions: Our results show that adolescent cocaine exposure modulates BDNF system early after treatment in the mesocorticolimbic pathway, identifying a complex but specific set of changes that could provide clues for treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/metabolism , Cocaine-Related Disorders/genetics , Nucleus Accumbens/metabolism , Animals , Cerebral Cortex/drug effects , Cocaine/administration & dosage , Male , Nucleus Accumbens/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, Opioid, kappa/genetics , Signal Transduction , Substance Withdrawal Syndrome/genetics
10.
J Neurosci ; 38(42): 8956-8966, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30185459

ABSTRACT

A single BDNF microinfusion into prelimbic (PrL) cortex immediately after the last cocaine self-administration session decreases relapse to cocaine-seeking. The BDNF effect is blocked by NMDAR antagonists. To determine whether synaptic activity in putative excitatory projection neurons in PrL cortex is sufficient for BDNF's effect on relapse, the PrL cortex of male rats was infused with an inhibitory Designer Receptor Exclusively Activated by Designer Drugs (DREADD) viral vector driven by an αCaMKII promoter. Immediately after the last cocaine self-administration session, rats were injected with clozapine-N-oxide 30 min before an intra-PrL BDNF microinfusion. DREADD-mediated inhibition of the PrL cortex blocked the BDNF-induced decrease in cocaine-seeking after abstinence and cue-induced reinstatement after extinction. Unexpectedly, DREADD inhibition of PrL neurons in PBS-infused rats also reduced cocaine-seeking, suggesting that divergent PrL pathways affect relapse. Next, using a cre-dependent retroviral approach, we tested the ability of DREADD inhibition of PrL projections to the NAc core or the paraventricular thalamic nucleus (PVT) to alter cocaine-seeking in BDNF- and PBS-infused rats. Selective inhibition of the PrL-NAc pathway at the end of cocaine self-administration blocked the BDNF-induced decrease in cocaine-seeking but had no effect in PBS-infused rats. In contrast, selective inhibition of the PrL-PVT pathway in PBS-infused rats decreased cocaine-seeking, and this effect was prevented in BDNF-infused rats. Thus, activity in the PrL-NAc pathway is responsible for the therapeutic effect of BDNF on cocaine-seeking whereas inhibition of activity in the PrL-pPVT pathway elicits a similar therapeutic effect in the absence of BDNF.SIGNIFICANCE STATEMENT The major issue in cocaine addiction is the high rate of relapse. However, the neuronal pathways governing relapse remain unclear. Using a pathway-specific chemogenetic approach, we found that BDNF differentially regulates two key prelimbic pathways to guide long-term relapse. Infusion of BDNF in the prelimbic cortex during early withdrawal from cocaine self-administration decreases relapse that is prevented when neurons projecting from the prelimbic cortex to the nucleus accumbens core are inhibited. In contrast, BDNF restores relapse when neurons projecting from the prelimbic cortex to the posterior paraventricular thalamic nucleus are inhibited. This study demonstrates that two divergent cortical outputs mediate relapse that is regulated in opposite directions by infusing BDNF in the prelimbic cortex during early withdrawal from cocaine.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Neurons/physiology , Prefrontal Cortex/physiology , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Clozapine/administration & dosage , Clozapine/analogs & derivatives , Drug-Seeking Behavior/drug effects , Male , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/physiology , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley
11.
Mol Neurobiol ; 55(7): 5453-5461, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28948570

ABSTRACT

Modulation of αCaMKII expression and phosphorylation is a feature shared by drugs of abuse with different mechanisms of action. Accordingly, we investigated whether αCaMKII expression and activation could be altered by self-administration of ketamine, a non-competitive antagonist of the NMDA glutamate receptor, with antidepressant and psychotomimetic as well as reinforcing properties. Rats self-administered ketamine at a sub-anesthetic dose for 43 days and were sacrificed 24 h after the last drug exposure; reward-related brain regions, such as medial prefrontal cortex (PFC), ventral striatum (vS), and hippocampus (Hip), were used for the measurement of αCaMKII-mediated signaling. αCaMKII phosphorylation was increased in these brain regions suggesting that ketamine, similarly to other reinforcers, activates this kinase. We next measured the two main targets of αCaMKII, i.e., GluN2B (S1303) and GluA1 (S831), and found increased activation of GluN2B (S1303) together with reduced phosphorylation of GluA1 (S831). Since GluN2B, via inhibition of ERK, regulates the membrane expression of GluA1, we measured ERK2 phosphorylation in the crude synaptosomal fraction of these brain regions, which was significantly reduced suggesting that ketamine-induced phosphorylation of αCaMKII promotes GluN2B (S1303) phosphorylation that, in turn, inhibits ERK 2 signaling, an effect that results in reduced membrane expression and phosphorylation of GluA1. Taken together, our findings point to αCaMKII autophosphorylation as a critical signature of ketamine self-administration providing an intracellular mechanism to explain the different effects caused by αCaMKII autophosphorylation on the post-synaptic GluN2B- and GluA1-mediated functions. These data add ketamine to the list of drugs of abuse converging on αCaMKII to sustain their addictive properties.


Subject(s)
Affect , Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Ketamine/administration & dosage , Reward , Affect/drug effects , Animals , Brain/pathology , Hippocampus/metabolism , Hippocampus/pathology , Male , Models, Biological , Phosphorylation/drug effects , Rats, Sprague-Dawley , Self Administration , Synaptosomes/metabolism
12.
Neurotox Res ; 32(4): 594-602, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28646469

ABSTRACT

Since the mid-to-late 2000s, synthetic cathinones have gained popularity among drug users due to their psychostimulant effects greater than those produced by cocaine and amphetamine. Among them, 3,4-methylenedioxypyrovalerone (MDPV) and 1-phenyl-2-(pyrrolidin-1-yl)pentan-1-one (α-PVP) are ones of the most popular cathinones available in the clandestine market as "bath salts" or "fertilizers." Pre-clinical studies indicate that MDPV and α-PVP induced psychomotor stimulation, affected thermoregulation, and promoted reinforcing properties in rodents. However, a direct comparative analysis on the effects caused by MDPV and α-PVP on the behavior and neuronal activation in rodents is still lacking. Behavioral analyses revealed that both MDPV and α-PVP affect spontaneous and stimulated motor responses. In particular, MDPV showed a greater psychomotor effect than α-PVP in line with its higher potency in blocking the dopamine transporter (DAT). Notably, MDPV was found to be more effective than α-PVP in facilitating spontaneous locomotion and it displayed a biphasic effect in contrast to the monophasically stimulated locomotion induced by α-PVP. In addition to the behavioral results, we also found a different modulation of immediate early genes (IEGs) such as Arc/Arg3.1 and c-Fos in the frontal lobe, striatum, and hippocampus, indicating that these drugs do impact brain homeostasis with changes in neuronal activity that depend on the drug, the brain area analyzed, and the timing after the injection. These results provide the first discrimination between MDPV and α-PVP based on behavioral and molecular data that may contribute to explain, at least in part, their toxicity.


Subject(s)
Alkaloids/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Motor Activity/drug effects , Pentanones/pharmacology , Pyrrolidines/pharmacology , Alkaloids/administration & dosage , Animals , Central Nervous System Stimulants/pharmacology , Designer Drugs , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , Male , Mice , Pentanones/administration & dosage , Pyrrolidines/administration & dosage
13.
Psychopharmacology (Berl) ; 234(8): 1217-1222, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28204841

ABSTRACT

RATIONALE AND OBJECTIVES: Among the changes caused by repeated exposure to drugs of abuse, structural rearrangements play a critical role, setting the stage for maladaptive responses to environmental challenges and sustaining drug-taking and drug-seeking behaviors. Given that adolescents are more vulnerable to drug abuse than adults and based on our recent data showing that a single exposure to cocaine during adolescence is sufficient to change the adolescent brain, we decided to investigate whether acute cocaine exposure may alter actin remodeling in reward-related brain regions. METHODS: Accordingly, we decided to evaluate if F-actin/G-actin ratio was altered by a single injection of cocaine (20 mg/kg) at postnatal day 35. We also evaluated whether the first administration of cocaine influences such a ratio in response to a second injection (10 mg/kg) provided 24 h or 7 days later. RESULTS: Within the medial prefrontal cortex, a single cocaine injection increases the F-actin/G-actin ratio. This effect lasts 1 week, and it is not affected by the second injection of cocaine, indicating a persistent effect of the first exposure. In the nucleus accumbens, cocaine reduces the F-actin/G-actin ratio 24 h later. Seven days later, instead, such a ratio is markedly increased: notably, the additional exposure to the psychostimulant normalizes the F-actin/G-actin ratio. CONCLUSIONS: These results suggest that a single cocaine injection during adolescence causes possible changes in actin dynamics and influences the response to a second challenge of the psychostimulant, indicating that early cocaine priming might affect mechanisms regulating synaptic structural plasticity in specific brain regions.


Subject(s)
Actins/drug effects , Cocaine/pharmacology , Prefrontal Cortex/drug effects , Analysis of Variance , Animals , Central Nervous System Stimulants/pharmacology , Cocaine/administration & dosage , Cocaine-Related Disorders/metabolism , Disease Models, Animal , Male , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Reward
14.
Neurotox Res ; 31(2): 289-297, 2017 02.
Article in English | MEDLINE | ID: mdl-27832448

ABSTRACT

During adolescence, the medial prefrontal cortex (mPFC) is still developing. We have previously shown that developmental cocaine exposure alters mPFC's ability to cope with challenging events. In this manuscript, we exposed rats developmentally treated with cocaine to a novelty task and analyzed the molecular changes of mPFC. Rats were exposed to cocaine from post-natal day (PND) 28 to PND 42 and sacrificed at PND 43, immediately after the novel object recognition (NOR) test. Cocaine-treated rats spent more time exploring the novel object than saline-treated counterparts, suggesting an increased response to novelty. The messenger RNA (mRNA) and protein levels of the immediate early gene Arc/Arg3.1 were reduced in both infralimbic (IL) and prelimbic (PL) cortices highlighting a baseline reduction of mPFC neuronal activity as a consequence of developmental exposure to cocaine. Intriguingly, significant molecular changes were observed in the IL, but not PL, cortex in response to the combination of cocaine exposure and test such as a marked upregulation of both Arc/Arg3.1 mRNA and protein levels only in cocaine-treated rats. As for proteins, such increase was observed only in the post-synaptic density and not in the whole homogenate, suggesting psychostimulant-induced changes in trafficking of Arc/Arg3.1 or an increased local translation. Notably, the same profile of Arc/Arg3.1 was observed for post-synaptic density (PSD)-95 leading to the possibility that Arc/Arg3.1 and PSD-95 bridge together to promote aberrant synaptic connectivity in IL cortex following repeated exposure to cocaine during brain development.


Subject(s)
Cocaine/adverse effects , Cytoskeletal Proteins/biosynthesis , Limbic Lobe/metabolism , Nerve Tissue Proteins/biosynthesis , Prefrontal Cortex/metabolism , Recognition, Psychology/drug effects , Animals , Disks Large Homolog 4 Protein , Exploratory Behavior/drug effects , Intracellular Signaling Peptides and Proteins/biosynthesis , Limbic Lobe/drug effects , Male , Membrane Proteins/biosynthesis , Post-Synaptic Density/metabolism , Prefrontal Cortex/drug effects , Rats , Up-Regulation/drug effects
15.
Mol Neurobiol ; 54(9): 7186-7193, 2017 11.
Article in English | MEDLINE | ID: mdl-27796754

ABSTRACT

Ketamine is a non-competitive antagonist of the NMDA glutamate receptor with psychotomimetic and reinforcing properties, although recent work has pointed out its antidepressant action following acute exposure. Our aim was to investigate the expression of crucial components of the glutamate synapse following chronic ketamine self-administration (S/A), focusing our attention on medial prefrontal cortex (mPFC) and hippocampus (Hip), two brain regions involved in compulsive drug-seeking and drug-related cognitive disorders. Rats self-administered ketamine at a sub-anesthetic dose for 5-6 weeks and were sacrificed 24 h after the last drug exposure. We found a general downregulation of glutamate receptor expression that was brain region-dependent. In fact, in the mPFC, we found reduced expression of NMDA receptor subunits, whereas AMPA receptor protein levels were reduced in Hip; of note, specific scaffolding proteins of NMDA and AMPA receptors were also reduced in mPFC and Hip, respectively. Moreover, the metabotropic mGluR5 receptor was similarly downregulated in these brain regions. These findings reveal a dynamic impairment of glutamate homeostasis in the mPFC and Hip that may represent a signature of long-term exposure to ketamine S/A. Further, this decrement, similarly observed in humans and animal models of schizophrenia may represent a specific feature of the human disease endophenotype.


Subject(s)
Brain/metabolism , Excitatory Amino Acid Antagonists/administration & dosage , Glutamic Acid/metabolism , Homeostasis/physiology , Ketamine/administration & dosage , Synapses/metabolism , Animals , Brain/drug effects , Excitatory Amino Acid Antagonists/toxicity , Homeostasis/drug effects , Ketamine/toxicity , Male , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Self Administration , Synapses/drug effects
16.
J Neurosci ; 36(31): 8149-59, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27488635

ABSTRACT

UNLABELLED: Cocaine exposure alters brain-derived neurotrophic factor (BDNF) expression in the brain. BDNF signaling through TrkB receptors differentially modulates cocaine self-administration, depending on the brain regions involved. In the present study, we determined how brain-wide inhibition of TrkB signaling affects cocaine intake, the motivation for the drug, and reinstatement of drug taking after extinction. To overcome the inability of TrkB ligands to cross the blood-brain barrier, the TrkB antagonist cyclotraxin-B was fused to the nontoxic transduction domain of the tat protein from human immunodeficiency virus type 1 (tat-cyclotraxin-B). Intravenous injection of tat-cyclotraxin-B dose-dependently reduced cocaine intake, motivation for cocaine (as measured under a progressive ratio schedule of reinforcement), and reinstatement of cocaine taking in rats allowed either short or long access to cocaine self-administration. In contrast, the treatment did not affect operant responding for a highly palatable sweet solution, demonstrating that the effects of tat-cyclotraxin-B are specific for cocaine reinforcement. Cocaine self-administration increased TrkB signaling and activated the downstream Akt pathway in the nucleus accumbens, and had opposite effects in the prefrontal cortex. Pretreatment with tat-cyclotraxin-B normalized protein levels in these two dopamine-innervated brain regions. Cocaine self-administration also increased TrkB signaling in the ventral tegmental area, where the dopaminergic projections originate, but pretreatment with tat-cyclotraxin-B did not alter this effect. Altogether, our data show that systemic administration of a brain-penetrant TrkB antagonist leads to brain region-specific effects and may be a potential pharmacological strategy for the treatment of cocaine addiction. SIGNIFICANCE STATEMENT: Brain-derived neurotrophic factor (BDNF) signaling through TrkB receptors plays a well established role in cocaine reinforcement. However, local manipulation of BDNF signaling yields divergent effects, depending on the brain region, thereby questioning the viability of systemic TrkB targeting for the treatment of cocaine use disorders. Our study provides first-time evidence that systemic administration of a brain-penetrant TrkB antagonist (tat-cyclotraxin-B) reduces several behavioral measures of cocaine dependence, without altering motor performance or reinforcement by a sweet palatable solution. In addition, although cocaine self-administration produced opposite effects on TrkB signaling in the nucleus accumbens and prefrontal cortex, tat-cyclotraxin-B administration normalized these cocaine-induced changes in both brain regions.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Nucleus Accumbens/metabolism , Peptides, Cyclic/administration & dosage , Prefrontal Cortex/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Injections, Intravenous , Male , Membrane Glycoproteins/metabolism , Nucleus Accumbens/drug effects , Peptides, Cyclic/pharmacokinetics , Prefrontal Cortex/drug effects , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Wistar , Receptor, trkB , Self Administration/methods , Treatment Outcome
17.
Psychopharmacology (Berl) ; 233(17): 3149-60, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27370019

ABSTRACT

RATIONALE: Although brain-derived neurotrophic factor (BDNF) is part of a homeostatic pathway involved in the development of alcohol dependence, it is not clear whether this is also true after recreational ethanol consumption. OBJECTIVES: We examined BDNF expression and signaling in the cortico-striatal network immediately and 24 h after either a single intravenous (i.v.) ethanol operant self-administration session or the last of 14 sessions. METHODS: To compare contingent and non-contingent ethanol exposure, we incorporated the "yoked control-operant paradigm" in which rats actively taking ethanol (S-Et) were paired with two yoked controls receiving passive infusions of ethanol (Y-Et) or saline. RESULTS: A single ethanol exposure transiently reduced BDNF mRNA levels in the medial prefrontal cortex (mPFC) of Y-Et. Immediately after the last of 14 sessions, mRNA and mature BDNF protein levels (mBDNF) were reduced in the mPFC in both S-Et and Y-Et while mBDNF expression was raised in the nucleus accumbens (NAc), suggesting enhanced anterograde transport from the mPFC. Conversely, 24 h later mBDNF expression and signaling were raised in the mPFC and NAc of S-Et rats but reduced in the NAc of Y-Et rats, with concomitant reduction of downstream signaling pathways. CONCLUSIONS: Our findings indicate that recreational-like i.v. doses of ethanol promote early changes in neurotrophin expression, depending on the length and modality of administration, the brain region investigated, and the presence of the drug. A rapid intervention targeting the BDNF system might be useful to prevent escalation to alcohol abuse.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Central Nervous System Depressants/pharmacology , Conditioning, Operant , Ethanol/pharmacology , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , RNA, Messenger/drug effects , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Central Nervous System Depressants/administration & dosage , Ethanol/administration & dosage , Male , Neostriatum/drug effects , Neostriatum/metabolism , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Self Administration , Signal Transduction/drug effects
18.
Psychopharmacology (Berl) ; 233(14): 2699-704, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27129861

ABSTRACT

RATIONALE: Adolescence is a period of high vulnerability to drugs of abuse and alterations of the proper developmental trajectory via psychostimulant exposure might change the physiological brain homeostasis. OBJECTIVE: By microdissection of brain areas via punching, we investigated whether repeated exposure to cocaine during adolescence (from postnatal day 28 [PND28] to PND42) has altered fibroblast growth factor-2 (FGF-2) messenger RNA (mRNA) levels in selected brain subregions critical for the action of cocaine. RESULTS: We found a reduction of FGF-2 mRNA levels in ventral tegmental area (VTA), where mesocortical and mesolimbic pathways originate. The analysis of the trophic factor levels in the distal projecting regions revealed a selective reduction of FGF-2 mRNA levels in infralimbic (IL) subregion of the medial prefrontal cortex (the terminal region of the mesocortical pathway) and in the nucleus accumbens core (cNAc) (the terminal region of the mesolimbic pathway). Last, we found reduced FGF-2 mRNA levels also in brain regions which, although in a different manner, contribute to the reward system, i.e., the central nucleus of amygdala (cAmy) and the ventral portion of hippocampus (vHip). CONCLUSION: The widespread and coordinated reduction of FGF-2 mRNA levels across the brain's reward neurocircuitry might represent a defensive strategy set in motion to oppose to the psychostimulant properties of cocaine. Moreover, given the role of FGF-2 in modulating mood disorders, the reduced trophic support here observed might sustain the negative emotional state set in motion by repeated exposure to cocaine.


Subject(s)
Brain/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Fibroblast Growth Factor 2/biosynthesis , Amygdala/metabolism , Animals , Brain/metabolism , Fibroblast Growth Factor 2/metabolism , Hippocampus/drug effects , Male , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reward , Ventral Tegmental Area/metabolism
19.
Hippocampus ; 26(6): 700-4, 2016 06.
Article in English | MEDLINE | ID: mdl-26850084

ABSTRACT

We previously demonstrated that nELAV/GAP-43 pathway is pivotal for learning and its hippocampal expression is up-regulated by acute stress following repeated cocaine administration. We therefore hypothesized that abstinence-induced stress may sustain nELAV/GAP-43 pathway during early abstinence following 2 weeks of cocaine self-administration. We found that contingent, but not non-contingent, cocaine exposure selectively increases hippocampal nELAV, but not GAP-43, expression immediately after the last self-administration session, an effect that wanes after 24 h and that comes back 7 days later when nELAV activation becomes associated with increased expression of GAP-43, an effect again observed only in animals self-administering the psychostimulant. Such effect is specific for nELAV since the ubiquitous ELAV/HuR is unchanged. This nELAV profile suggests that its initial transient alteration is perhaps related to the daily administration of cocaine, while the increase in the nELAV/GAP-43 pathway following a week of abstinence may reflect the activation of this cascade as a target of stressful conditions associated with drug-related memories. © 2016 Wiley Periodicals, Inc.


Subject(s)
Central Nervous System Stimulants/administration & dosage , Cocaine/administration & dosage , GAP-43 Protein , Hippocampus/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Blotting, Western , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Disease Models, Animal , ELAV-Like Protein 1/metabolism , GAP-43 Protein/metabolism , Rats , Self Administration , Signal Transduction , Stress, Psychological/etiology , Stress, Psychological/metabolism
20.
Pharmacol Res ; 104: 22-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26706783

ABSTRACT

Ketamine is a drug of abuse with a unique profile, which besides its inherent mechanism of action as a non-competitive antagonist of the NMDA glutamate receptor, displays both antidepressant and reinforcing properties. The major aim of our study was to find a molecular signature of ketamine that may help in discriminating between its reinforcing and antidepressant effects. To this end, we focused our attention on BDNF, a neurotrophin that has been shown to play a role in both antidepressant and reinforcing properties of several drugs. Rats were exposed to self-administer intravenous (IV) ketamine (S/A) for 43 days or to receive a single IV ketamine 0.5mg/kg, or vehicle infusion. Although the dose we employed is lower than that reported by the literature, it however yields Cmax values that correspond to those achieved in humans after antidepressant treatment. Our results show that while the single infusion of ketamine increased the neurotrophin expression in the hippocampus while reducing it in the ventral striatum, a feature shared with other antidepressants, the repeated self-administration reduced mBDNF expression and its downstream signalling in both ventral striatum and hippocampus. Further, we here show that phosphorylation of Akt is oppositely regulated by ketamine, pointing to this pathway as central to the different actions of the drug. Taken together, we here point to BDNF and its downstream signalling pathway as a finely tuned mechanism whose modulation might subserve the different features of ketamine.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Ketamine/pharmacology , Reinforcement, Psychology , Animals , Antidepressive Agents/administration & dosage , Brain/metabolism , Early Growth Response Protein 1/metabolism , Infusions, Intravenous , Ketamine/administration & dosage , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Self Administration , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL