Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.601
Filter
Add more filters

Publication year range
1.
Cell ; 165(7): 1776-1788, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27238022

ABSTRACT

A major challenge in understanding the cellular diversity of the brain has been linking activity during behavior with standard cellular typology. For example, it has not been possible to determine whether principal neurons in prefrontal cortex active during distinct experiences represent separable cell types, and it is not known whether these differentially active cells exert distinct causal influences on behavior. Here, we develop quantitative hydrogel-based technologies to connect activity in cells reporting on behavioral experience with measures for both brain-wide wiring and molecular phenotype. We find that positive and negative-valence experiences in prefrontal cortex are represented by cell populations that differ in their causal impact on behavior, long-range wiring, and gene expression profiles, with the major discriminant being expression of the adaptation-linked gene NPAS4. These findings illuminate cellular logic of prefrontal cortex information processing and natural adaptive behavior and may point the way to cell-type-specific understanding and treatment of disease-associated states.


Subject(s)
Behavior, Animal , Brain Mapping/methods , Prefrontal Cortex/cytology , Animals , Appetitive Behavior , Basic Helix-Loop-Helix Transcription Factors/genetics , Cocaine/administration & dosage , Electroshock , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prefrontal Cortex/metabolism
2.
Cell ; 152(1-2): 236-47, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332758

ABSTRACT

The sigma-1 receptor (Sig-1R), an endoplasmic reticulum (ER) chaperone protein, is an interorganelle signaling modulator that potentially plays a role in drug-seeking behaviors. However, the brain site of action and underlying cellular mechanisms remain unidentified. We found that cocaine exposure triggers a Sig-1R-dependent upregulation of D-type K(+) current in the nucleus accumbens (NAc) that results in neuronal hypoactivity and thereby enhances behavioral cocaine response. Combining ex vivo and in vitro studies, we demonstrated that this neuroadaptation is caused by a persistent protein-protein association between Sig-1Rs and Kv1.2 channels, a phenomenon that is associated to a redistribution of both proteins from intracellular compartments to the plasma membrane. In conclusion, the dynamic Sig-1R-Kv1.2 complex represents a mechanism that shapes neuronal and behavioral response to cocaine. Functional consequences of Sig-1R binding to K(+) channels may have implications for other chronic diseases where maladaptive intrinsic plasticity and Sig-1Rs are engaged.


Subject(s)
Cocaine/administration & dosage , Kv1.2 Potassium Channel/metabolism , Neuronal Plasticity , Nucleus Accumbens/metabolism , Receptors, sigma/metabolism , Animals , Drug-Seeking Behavior , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Receptors, sigma/genetics , Sigma-1 Receptor
3.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38485256

ABSTRACT

The ventral pallidum (VP) is a central hub in the reward circuitry with diverse projections that have different behavioral roles attributed mostly to the connectivity with the downstream target. However, different VP projections may represent, as in the striatum, separate neuronal populations that differ in more than just connectivity. In this study, we performed in mice of both sexes a multimodal dissection of four major projections of the VP-to the lateral hypothalamus (VP→LH), ventral tegmental area (VP→VTA), lateral habenula (VP→LHb), and mediodorsal thalamus (VP→MDT)-with physiological, anatomical, genetic, and behavioral tools. We also tested for physiological differences between VP neurons receiving input from nucleus accumbens medium spiny neurons (MSNs) that express either the D1 (D1-MSNs) or the D2 (D2-MSNs) dopamine receptor. We show that each VP projection (1) when inhibited during a cocaine conditioned place preference (CPP) test affects performance differently, (2) receives a different pattern of inputs using rabies retrograde labeling, (3) shows differentially expressed genes using RNA sequencing, and (4) has projection-specific characteristics in excitability and synaptic input characteristics using whole-cell patch clamp. VP→LH and VP→VTA projections have different effects on CPP and show low overlap in circuit tracing experiments, as VP→VTA neurons receive more striatal input, while VP→LH neurons receive more olfactory input. Additionally, VP→VTA neurons are less excitable, while VP→LH neurons are more excitable than the average VP neuron, a difference driven mainly by D2-MSN-responding neurons. Thus, VP→VTA and VP→LH neurons may represent largely distinct populations of VP neurons.


Subject(s)
Basal Forebrain , Cocaine , Neural Pathways , Reward , Animals , Mice , Basal Forebrain/physiology , Male , Cocaine/pharmacology , Cocaine/administration & dosage , Female , Neural Pathways/physiology , Mice, Inbred C57BL , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D2/genetics , Ventral Tegmental Area/physiology , Ventral Tegmental Area/cytology
4.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38719446

ABSTRACT

Drugs of abuse cause changes in the prefrontal cortex (PFC) and associated regions that impair inhibitory control over drug-seeking. Breaking the contingencies between drug-associated cues and the delivery of the reward during extinction learning reduces relapse. Vagus nerve stimulation (VNS) has previously been shown to enhance extinction learning and reduce drug-seeking. Here we determined the effects of VNS-mediated release of brain-derived neurotrophic factor (BDNF) on extinction and cue-induced reinstatement in male rats trained to self-administer cocaine. Pairing 10 d of extinction training with VNS facilitated extinction and reduced drug-seeking behavior during reinstatement. Rats that received a single extinction session with VNS showed elevated BDNF levels in the medial PFC as determined via an enzyme-linked immunosorbent assay. Systemic blockade of tropomyosin receptor kinase B (TrkB) receptors during extinction, via the TrkB antagonist ANA-12, decreased the effects of VNS on extinction and reinstatement. Whole-cell recordings in brain slices showed that cocaine self-administration induced alterations in the ratio of AMPA and NMDA receptor-mediated currents in Layer 5 pyramidal neurons of the infralimbic cortex (IL). Pairing extinction with VNS reversed cocaine-induced changes in glutamatergic transmission by enhancing AMPAR currents, and this effect was blocked by ANA-12. Our study suggests that VNS consolidates the extinction of drug-seeking behavior by reversing drug-induced changes in synaptic AMPA receptors in the IL, and this effect is abolished by blocking TrkB receptors during extinction, highlighting a potential mechanism for the therapeutic effects of VNS in addiction.


Subject(s)
Drug-Seeking Behavior , Extinction, Psychological , Neuronal Plasticity , Prefrontal Cortex , Rats, Sprague-Dawley , Receptor, trkB , Vagus Nerve Stimulation , Animals , Male , Rats , Vagus Nerve Stimulation/methods , Drug-Seeking Behavior/physiology , Drug-Seeking Behavior/drug effects , Receptor, trkB/metabolism , Receptor, trkB/antagonists & inhibitors , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Extinction, Psychological/physiology , Extinction, Psychological/drug effects , Prefrontal Cortex/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Self Administration , Cocaine/pharmacology , Cocaine/administration & dosage
5.
J Neurosci ; 44(17)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38514181

ABSTRACT

The initiation of abstinence after chronic drug self-administration is stressful. Cocaine-seeking behavior on the first day of the absence of the expected drug (Extinction Day 1, ED1) is reduced by blocking 5-HT signaling in dorsal hippocampal cornu ammonis 1 (CA1) in both male and female rats. We hypothesized that the experience of ED1 can substantially influence later relapse behavior and that dorsal raphe (DR) serotonin (5-HT) input to CA1 may be involved. We inhibited 5-HT1A/1B receptors (WAY-100635 plus GR-127935), or DR input (chemogenetics), in CA1 on ED1 to test the role of this pathway on cocaine-seeking persistence 2 weeks later. We also inhibited 5-HT1A or 5-HT1B receptors in CA1 during conditioned place preference (CPP) for cocaine, to examine mechanisms involved in the persistent effects of ED1 manipulations. Inhibition of DR inputs, or 5-HT1A/1B signaling, in CA1 decreased drug seeking on ED1 and decreased cocaine seeking 2 weeks later revealing that 5-HT signaling in CA1 during ED1 contributes to persistent drug seeking during abstinence. In addition, 5-HT1B antagonism alone transiently decreased drug-associated memory performance when given prior to a CPP test, whereas similar antagonism of 5-HT1A alone had no such effect but blocked CPP retrieval on a test 24 h later. These CPP findings are consistent with prior work showing that DR inputs to CA1 augment recall of the drug-associated context and drug seeking via 5-HT1B receptors and prevent consolidation of the updated nondrug context via 5-HT1A receptors. Thus, treatments that modulate 5-HT-dependent memory mechanisms in CA1 during initial abstinence may facilitate later maintenance of abstinence.


Subject(s)
Cocaine , Drug-Seeking Behavior , Oxadiazoles , Serotonin , Animals , Male , Drug-Seeking Behavior/physiology , Drug-Seeking Behavior/drug effects , Rats , Serotonin/metabolism , Female , Cocaine/administration & dosage , Cocaine/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Pyridines/pharmacology , Serotonin Antagonists/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Piperazines/pharmacology , Rats, Sprague-Dawley , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Self Administration , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Receptor, Serotonin, 5-HT1B/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism
6.
Behav Pharmacol ; 35(4): 147-155, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38651979

ABSTRACT

Previous exposure to drugs of abuse produces impairments in studies of reversal learning, delay discounting and response inhibition tasks. While these studies contribute to the understanding of normal decision-making and how it is impaired by drugs of abuse, they do not fully capture how decision-making impacts the ability to delay gratification for greater long-term benefit. To address this issue, we used a diminishing returns task to study decision-making in rats that had previously self-administered cocaine. This task was designed to test the ability of the rat to choose to delay gratification in the short-term to obtain more reward over the course of the entire behavioral session. Rats were presented with two choices. One choice had a fixed amount of time delay needed to obtain reward [i.e. fixed delay (FD)], while the other choice had a progressive delay (PD) that started at 0 s and progressively increased by 1 s each time the PD option was selected. During the 'reset' variation of the task, rats could choose the FD option to reset the time delay associated with the PD option. Consistent with previous results, we found that prior cocaine exposure reduced rats' overall preference for the PD option in post-task reversal testing during 'no-reset' sessions, suggesting that cocaine exposure made rats more sensitive to the increasing delay of the PD option. Surprisingly, however, we found that rats that had self-administered cocaine 1-month prior, adapted behavior during 'reset' sessions by delaying gratification to obtain more reward in the long run similar to control rats.


Subject(s)
Cocaine , Delay Discounting , Reward , Self Administration , Animals , Cocaine/pharmacology , Cocaine/administration & dosage , Male , Delay Discounting/drug effects , Rats , Choice Behavior/drug effects , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Decision Making/drug effects , Cocaine-Related Disorders/psychology , Rats, Long-Evans , Time Factors
7.
Dev Psychobiol ; 66(6): e22514, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38922890

ABSTRACT

Repeated exposure to abused drugs leads to reorganizing synaptic connections in the brain, playing a pivotal role in the relapse process. Additionally, recent research has highlighted the impact of parental drug exposure before gestation on subsequent generations. This study aimed to explore the influence of parental morphine exposure 10 days prior to pregnancy on drug-induced locomotor sensitization. Adult male and female Wistar rats were categorized into morphine-exposed and control groups. Ten days after their last treatment, they were mated, and their male offspring underwent morphine, methamphetamine, cocaine, and nicotine-induced locomotor sensitization tests. The results indicated increased locomotor activity in both groups after drug exposure, although the changes were attenuated in morphine and cocaine sensitization among the offspring of morphine-exposed parents (MEPs). Western blotting analysis revealed altered levels of D2 dopamine receptors (D2DRs) in the prefrontal cortex and nucleus accumbens of the offspring from MEPs. Remarkably, despite not having direct in utero drug exposure, these offspring exhibited molecular alterations affecting morphine and cocaine-induced sensitization. The diminished sensitization to morphine and cocaine suggested the development of a tolerance phenotype in these offspring. The changes in D2DR levels in the brain might play a role in these adaptations.


Subject(s)
Cocaine , Locomotion , Morphine , Nucleus Accumbens , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Rats, Wistar , Receptors, Dopamine D2 , Animals , Female , Morphine/pharmacology , Morphine/administration & dosage , Male , Cocaine/pharmacology , Cocaine/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/chemically induced , Rats , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D2/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Locomotion/drug effects , Behavior, Animal/drug effects , Behavior, Animal/physiology , Narcotics/pharmacology , Paternal Exposure/adverse effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Motor Activity/drug effects , Motor Activity/physiology
8.
J Neurosci ; 41(5): 1046-1058, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33268545

ABSTRACT

Substance use disorder (SUD) is associated with disruptions in circadian rhythms. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration (acquisition, dose-response, progressive ratio, extinction, cue-induced reinstatement) in wild-type (WT) and Npas2 mutant mice at different times of day. In the light (inactive) phase, cocaine self-administration, reinforcement, motivation and extinction responding were increased in all Npas2 mutants. Sex differences emerged during the dark (active) phase with Npas2 mutation increasing self-administration, extinction responding, and reinstatement only in females as well as reinforcement and motivation in males and females. To determine whether circulating hormones are driving these sex differences, we ovariectomized WT and Npas2 mutant females and confirmed that unlike sham controls, ovariectomized mutant mice showed no increase in self-administration. To identify whether striatal brain regions are activated in Npas2 mutant females, we measured cocaine-induced ΔFosB expression. Relative to WT, ΔFosB expression was increased in D1+ neurons in the nucleus accumbens (NAc) core and dorsolateral (DLS) striatum in Npas2 mutant females after dark phase self-administration. We also identified potential target genes that may underlie the behavioral responses to cocaine in Npas2 mutant females. These results suggest NPAS2 regulates reward and activity in specific striatal regions in a sex and time of day (TOD)-specific manner. Striatal activation could be augmented by circulating sex hormones, leading to an increased effect of Npas2 mutation in females.SIGNIFICANCE STATEMENT Circadian disruptions are a common symptom of substance use disorders (SUDs) and chronic exposure to drugs of abuse alters circadian rhythms, which may contribute to subsequent SU. Diurnal rhythms are commonly found in behavioral responses to drugs of abuse with drug sensitivity and motivation peaking during the dark (active) phase in nocturnal rodents. Emerging evidence links disrupted circadian genes to SU vulnerability and drug-induced alterations to these genes may augment drug-seeking. The circadian transcription factor neuronal PAS domain protein 2 (NPAS2) is enriched in reward-related brain regions and regulates reward, but its role in SU is unclear. To examine the role of NPAS2 in drug taking, we measured intravenous cocaine self-administration in wild-type (WT) and Npas2 mutant mice at different times of day.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Circadian Rhythm/physiology , Cocaine/administration & dosage , Mutation/genetics , Nerve Tissue Proteins/genetics , Sex Characteristics , Administration, Intravenous , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Circadian Rhythm/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Self Administration
9.
J Neurosci ; 41(39): 8262-8277, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34413203

ABSTRACT

Cue-induced cocaine craving progressively intensifies (incubates) after withdrawal from cocaine self-administration in rats and humans. In rats, the expression of incubation ultimately depends on Ca2+-permeable AMPARs that accumulate in synapses onto medium spiny neurons (MSNs) in the NAc core. However, the delay in their accumulation (∼1 month after drug self-administration ceases) suggests earlier waves of plasticity. This prompted us to conduct the first study of NMDAR transmission in NAc core during incubation, focusing on the GluN3 subunit, which confers atypical properties when incorporated into NMDARs, including insensitivity to Mg2+ block and Ca2+ impermeability. Whole-cell patch-clamp recordings were conducted in MSNs of adult male rats 1-68 d after discontinuing extended-access saline or cocaine self-administration. NMDAR transmission was enhanced after 5 d of cocaine withdrawal, and this persisted for at least 68 d of withdrawal. The earliest functional alterations were mediated through increased contributions of GluN2B-containing NMDARs, followed by increased contributions of GluN3-containing NMDARs. As predicted by GluN3-NMDAR incorporation, fewer MSN spines exhibited NMDAR-mediated Ca2+ entry. GluN3A knockdown in NAc core was sufficient to prevent incubation of craving, consistent with biotinylation studies showing increased GluN3A surface expression, although array tomography studies suggested that adaptations involving GluN3B also occur. Collectively, our data show that a complex cascade of NMDAR and AMPAR plasticity occurs in NAc core, potentially through a homeostatic mechanism, leading to persistent increases in cocaine cue reactivity and relapse vulnerability. This is a remarkable example of experience-dependent glutamatergic plasticity evolving over a protracted window in the adult brain.SIGNIFICANCE STATEMENT "Incubation of craving" is an animal model for the persistence of vulnerability to cue-induced relapse after prolonged drug abstinence. Incubation also occurs in human drug users. AMPAR plasticity in medium spiny neurons (MSNs) of the NAc core is critical for incubation of cocaine craving but occurs only after a delay. Here we found that AMPAR plasticity is preceded by NMDAR plasticity that is essential for incubation and involves GluN3, an atypical NMDAR subunit that markedly alters NMDAR transmission. Together with AMPAR plasticity, this represents profound remodeling of excitatory synaptic transmission onto MSNs. Given the importance of MSNs for translating motivation into action, this plasticity may explain, at least in part, the profound shifts in motivated behavior that characterize addiction.


Subject(s)
Cocaine/administration & dosage , Craving/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Membrane Glycoproteins/metabolism , Nucleus Accumbens/metabolism , Animals , Calcium/metabolism , Drug-Seeking Behavior/physiology , Male , Nucleus Accumbens/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Self Administration
10.
J Neurosci ; 41(13): 2814-2827, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33602824

ABSTRACT

Epigenetic mechanisms regulate processes of neuroplasticity critical to cocaine-induced behaviors. This includes the Class I histone deacetylase (HDAC) HDAC3, known to act as a negative regulator of cocaine-associated memory formation within the nucleus accumbens (NAc). Despite this, it remains unknown how cocaine alters HDAC3-dependent mechanisms. Here, we profiled HDAC3 expression and activity in total NAc mouse tissue following cocaine exposure. Although chronic cocaine did not affect expression of Hdac3 within the NAc, chronic cocaine did affect promoter-specific changes in HDAC3 and H4K8Ac occupancy. These changes in promoter occupancy correlated with cocaine-induced changes in expression of plasticity-related genes. To causally determine whether cocaine-induced plasticity is mediated by HDAC3's deacetylase activity, we overexpressed a deacetylase-dead HDAC3 point mutant (HDAC3-Y298H-v5) within the NAc of adult male mice. We found that disrupting HDAC3's enzymatic activity altered selective changes in gene expression and synaptic plasticity following cocaine exposure, despite having no effects on cocaine-induced behaviors. In further assessing HDAC3's role within the NAc, we observed that chronic cocaine increases Hdac3 expression in Drd1 but not Drd2-cells of the NAc. Moreover, we discovered that HDAC3 acts selectively within D1R cell-types to regulate cocaine-associated memory formation and cocaine-seeking. Overall, these results suggest that cocaine induces cell-type-specific changes in epigenetic mechanisms to promote plasticity important for driving cocaine-related behaviors.SIGNIFICANCE STATEMENT Drugs of abuse alter molecular mechanisms throughout the reward circuitry that can lead to persistent drug-associated behaviors. Epigenetic regulators are critical drivers of drug-induced changes in gene expression. Here, we demonstrate that the activity of an epigenetic enzyme promotes neuroplasticity within the nucleus accumbens (NAc) critical to cocaine action. In addition, we demonstrate that these changes in epigenetic activity drive cocaine-seeking behaviors in a cell-type-specific manner. These findings are key in understanding and targeting cocaine's impact of neural circuitry and behavior.


Subject(s)
Cocaine/administration & dosage , Drug-Seeking Behavior/physiology , Histone Deacetylases/biosynthesis , Neuronal Plasticity/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/enzymology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dopamine Uptake Inhibitors/administration & dosage , Drug-Seeking Behavior/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/physiology , Histone Deacetylases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Self Administration
11.
J Biol Chem ; 297(6): 101361, 2021 12.
Article in English | MEDLINE | ID: mdl-34756883

ABSTRACT

The dopamine (DA) transporter (DAT) is part of a presynaptic multiprotein network involving interactions with scaffold proteins via its C-terminal PDZ domain-binding sequence. Using a mouse model expressing DAT with mutated PDZ-binding sequence (DAT-AAA), we previously demonstrated the importance of this binding sequence for striatal expression of DAT. Here, we show by application of direct stochastic reconstruction microscopy not only that the striatal level of transporter is reduced in DAT-AAA mice but also that the nanoscale distribution of this transporter is altered with a higher propensity of DAT-AAA to localize to irregular nanodomains in dopaminergic terminals. In parallel, we observe mesostriatal DA adaptations and changes in DA-related behaviors distinct from those seen in other genetic DAT mouse models. DA levels in the striatum are reduced to ∼45% of that of WT, accompanied by elevated DA turnover. Nonetheless, fast-scan cyclic voltammetry recordings on striatal slices reveal a larger amplitude and prolonged clearance rate of evoked DA release in DAT-AAA mice compared with WT mice. Autoradiography and radioligand binding show reduced DA D2 receptor levels, whereas immunohistochemistry and autoradiography show unchanged DA D1 receptor levels. In behavioral experiments, we observe enhanced self-administration of liquid food under both a fixed ratio of one and progressive ratio schedule of reinforcement but a reduction compared with WT when using cocaine as reinforcer. In summary, our data demonstrate how disruption of PDZ domain interactions causes changes in DAT expression and its nanoscopic distribution that in turn alter DA clearance dynamics and related behaviors.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Homeostasis , Motivation , PDZ Domains , Reward , Animals , Binding Sites , Cocaine/administration & dosage , Conditioning, Operant , Male , Mice , Protein Binding , Self Administration
12.
Cereb Cortex ; 31(4): 1927-1952, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33253368

ABSTRACT

The hippocampus plays a key role in contextual conditioning and has been proposed as an important component of the cocaine addiction brain circuit. To gain knowledge about cocaine-induced alterations in this circuit, we used focused ion beam milling/scanning electron microscopy to reveal and quantify the three-dimensional synaptic organization of the neuropil of the stratum radiatum of the rat CA1, under normal circumstances and after cocaine-self administration (SA). Most synapses are asymmetric (excitatory), macular-shaped, and in contact with dendritic spine heads. After cocaine-SA, the size and the complexity of the shape of both asymmetric and symmetric (inhibitory) synapses increased but no changes were observed in the synaptic density. This work constitutes the first detailed report on the 3D synaptic organization in the stratum radiatum of the CA1 field of cocaine-SA rats. Our data contribute to the elucidation of the normal and altered synaptic organization of the hippocampus, which is crucial for better understanding the neurobiological mechanisms underlying cocaine addiction.


Subject(s)
CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/ultrastructure , Cocaine/administration & dosage , Imaging, Three-Dimensional/methods , Synapses/drug effects , Synapses/ultrastructure , Animals , CA1 Region, Hippocampal/pathology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dopamine Uptake Inhibitors/administration & dosage , Male , Microscopy, Electron, Scanning/methods , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Rats , Rats, Inbred Lew , Self Administration/methods , Synapses/pathology
13.
Eur J Neurosci ; 54(3): 4920-4933, 2021 08.
Article in English | MEDLINE | ID: mdl-34133054

ABSTRACT

The fragile X mental retardation protein (FMRP), an RNA-binding protein, regulates cocaine-induced neuronal plasticity and is critical for the normal development of drug-induced locomotor sensitization, as well as reward-related learning in the conditioned place preference assay. However, it is unknown whether FMRP impacts behaviors that are used to more closely model substance use disorders. Utilizing a cocaine intravenous self-administration (IVSA) assay in Fmr1 knockout (KO) and wild-type (WT) littermate mice, we find that, despite normal acquisition and extinction learning, Fmr1 KO mice fail to make a normal upward shift in responding during dose-response testing. Later, when given access to the original acquisition dose under increasing fixed ratio (FR) schedules of reinforcement (FR1, FR3, and FR5), Fmr1 KO mice earn significantly fewer cocaine infusions than WT mice. Importantly, similar deficits are not present in operant conditioning using a palatable food reinforcer, indicating that our results do not represent broad learning or reward-related deficits in Fmr1 KO mice. Additionally, we find an FMRP target, the activity-regulated cytoskeleton-associated protein (Arc), to be significantly reduced in synaptic cellular fractions prepared from the nucleus accumbens of Fmr1 KO, compared with WT, mice following operant tasks reinforced with cocaine but not food. Overall, our findings suggest that FMRP facilitates adjustments in drug self-administration behavior that generally serve to preserve reinforcement level, and combined with our similar IVSA findings in Arc KO mice may implicate Arc, along with FMRP, in behavioral shifts that occur in drug taking when drug availability is altered.


Subject(s)
Cocaine , Fragile X Mental Retardation Protein , Neuronal Plasticity , Animals , Cocaine/administration & dosage , Fragile X Mental Retardation Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Reinforcement, Psychology
14.
J Pharmacol Exp Ther ; 379(2): 108-116, 2021 11.
Article in English | MEDLINE | ID: mdl-34413199

ABSTRACT

A subset of rats that self-administer 3,4-methylenedioxypyrovalerone (MDPV) develop unusually high levels of drug taking. A history of responding maintained by cocaine, but not food, prevents the development of this high-responder phenotype; however, it is unclear how histories of noncontingent cocaine exposure or self-administering drugs from other pharmacological classes would affect its development. In the current studies, 5 groups of male Sprague-Dawley rats were used to determine whether histories of responding maintained by drugs from different pharmacological classes (e.g., MDPV, cocaine, fentanyl, nicotine, or ketamine) would differentially impact the development of the high-responder phenotype when MDPV was available for self-administration. Two additional groups were used to determine whether noncontingent exposure to cocaine would prevent the development of the high-responder phenotype when MDPV was available for self-administration, and whether noncontingent exposure to MDPV would facilitate the development of the high-responder phenotype when cocaine was available for self-administration. Consistent with previous reports, a history of response-contingent cocaine, and to a lesser extent noncontingent cocaine, prevented the MDPV high-responder phenotype; however, when responding was initially maintained by fentanyl, nicotine, or ketamine, the MDPV high-responder phenotype developed in ∼45% of rats. By manipulating behavioral and pharmacological histories prior to evaluating MDPV self-administration, the current studies provide additional evidence that a history of response-contingent (or noncontingent) cocaine can prevent the transition from well regulated to aberrant drug-taking when responding is maintained by MDPV. Although the mechanism(s) that underlies this novel high-responder phenotype are unknown, elucidation may provide insight into individual differences relating to substance use disorder. SIGNIFICANCE STATEMENT: A subset of outbred Sprague-Dawley rats self-administer high levels of the synthetic cathinone 3,4-methylenedioxypyrovalerone (MDPV). Understanding the behavioral and/or pharmacological factors that can prevent the development of dysregulated MDPV self-administration may provide insight into individual differences in vulnerability to develop a substance use disorder.


Subject(s)
Behavior, Addictive/psychology , Benzodioxoles/administration & dosage , Pyrrolidines/administration & dosage , Reinforcement Schedule , Adrenergic Uptake Inhibitors/administration & dosage , Animals , Behavior, Addictive/genetics , Cocaine/administration & dosage , Fentanyl/administration & dosage , Ketamine/administration & dosage , Male , Nicotine/administration & dosage , Rats , Rats, Sprague-Dawley , Self Administration/psychology , Synthetic Cathinone
15.
J Pharmacol Exp Ther ; 379(1): 12-19, 2021 10.
Article in English | MEDLINE | ID: mdl-34272270

ABSTRACT

Although dopamine plays a prominent role in mediating cocaine's abuse-related effects, the specific roles of dopamine receptor subtypes are not fully understood. Whereas the effects of drugs acting at dopamine D2-like receptors (D2Rs) have been characterized, less is known about dopamine D1-like receptors (D1Rs). The present experiments examined the effects of drugs with varying intrinsic efficacy at D1R on the relative reinforcing strength of cocaine in male cynomolgus monkeys. Use of socially housed monkeys permitted the assessment of whether social status influenced the behavioral effects of D1R-acting drugs. The high-efficacy D1R agonist SKF 81297, low-efficacy D1R agonist SKF 38393, and D1R antagonist SCH 23390 were administered acutely to monkeys self-administering cocaine under a food-cocaine choice procedure in which a cocaine-choice dose-effect curve was determined daily. To assess selectivity of behavioral effects on cocaine choice, effects of doses that did not disrupt responding (indicated by a ≥35% decrease in total reinforcers delivered) were analyzed. Neither SKF 81297 nor SCH 23390 affected cocaine choice in dominant or subordinate monkeys. However, the low-efficacy agonist SKF 38393 selectively decreased cocaine choice; this effect was larger and only reached statistical significance in subordinate monkeys. Increasing the time between D1-acting drug administration and the cocaine choice session did not affect these results. The results indicate that, like D2R-acting drugs, the behavioral effects of D1R-acting drugs on cocaine choice can vary according to intrinsic efficacy and social status. Moreover, they demonstrate that D1R-acting drugs affect behavior under a narrower range of conditions than D2R-acting drugs. SIGNIFICANCE STATEMENT: Cocaine use disorder represents an insidious public health concern with no Food and Drug Administration-approved medications. Although dopamine receptors have been strongly implicated in mediating the abuse-related effects of cocaine, the roles of dopamine receptor subtypes are incompletely understood. The present study in nonhuman primates found that cocaine choice was decreased only by a low-efficacy D1R agonist, and that this effect depended on the social status of the monkey.


Subject(s)
Choice Behavior/drug effects , Cocaine/administration & dosage , Dopamine Agonists/pharmacology , Food Preferences/drug effects , Receptors, Dopamine D1/agonists , Social Interaction/drug effects , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Choice Behavior/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dose-Response Relationship, Drug , Food Preferences/physiology , Food Preferences/psychology , Ligands , Macaca fascicularis , Male , Receptors, Dopamine D1/metabolism
16.
Nat Rev Neurosci ; 17(6): 351-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27150400

ABSTRACT

Although it is challenging for individuals with cocaine addiction to achieve abstinence, the greatest difficulty is avoiding relapse to drug taking, which is often triggered by cues associated with prior cocaine use. This vulnerability to relapse persists for long periods (months to years) after abstinence is achieved. Here, I discuss rodent studies of cue-induced cocaine craving during abstinence, with a focus on neuronal plasticity in the reward circuitry that maintains high levels of craving. Such work has the potential to identify new therapeutic targets and to further our understanding of experience-dependent plasticity in the adult brain under normal circumstances and in the context of addiction.


Subject(s)
Brain/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Craving/physiology , Nerve Net/metabolism , Synapses/metabolism , Animals , Behavior, Addictive/metabolism , Brain/drug effects , Craving/drug effects , Humans , Nerve Net/drug effects , Neuronal Plasticity/physiology , Reward , Synapses/drug effects
17.
Int J Neuropsychopharmacol ; 24(6): 499-504, 2021 07 14.
Article in English | MEDLINE | ID: mdl-33305794

ABSTRACT

The chronic relapsing nature of cocaine addiction suggests that chronic cocaine exposure produces persistent neuroadaptations that may be temporally and regionally dynamic in brain areas such as the dopaminergic (DA) system. We have previously shown altered metabolism of DA-target structures, the ventral and dorsal striatum, between early and late abstinence. However, specific changes within the midbrain DA system were not investigated. Here, we investigated potential time- and region-specific changes of activity in the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNc) in rats that had extended or limited access to cocaine and later underwent a period of abstinence. We found that DA activity is decreased only in the VTA in rats with extended access to cocaine, with no changes in SNc DA activity. These changes in VTA DA activity may participate in the negative emotional state and the incubation of drug seeking that occur during abstinence from cocaine.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Dopamine/metabolism , Pars Compacta/metabolism , Ventral Tegmental Area/metabolism , Animals , Behavior, Animal/physiology , Male , Rats , Rats, Sprague-Dawley , Self Administration
18.
FASEB J ; 34(7): 9192-9206, 2020 07.
Article in English | MEDLINE | ID: mdl-32421249

ABSTRACT

Maternal diet significantly influences the proper development of offspring in utero. Modifications of diet composition may lead to metabolic and mental disorders that may predispose offspring to a substance use disorder. We assessed the impact of a maternal high-sugar diet (HSD, rich in sucrose) consumed during pregnancy and lactation on the offspring phenotype in the context of the rewarding and motivational effects of cocaine and changes within the central melanocortin (MC) system. Using an intravenous cocaine self-administration model, we showed that maternal HSD leads to increased relapse of cocaine-seeking behavior in male offspring. In addition, we demonstrated that cocaine induces changes in the level of MC-4 receptors in the offspring brain, and these changes depend on maternal diet. These studies also reveal that an MC-4 receptor antagonist reduces the reinstatement of cocaine-seeking behavior, and offspring exposed to maternal HSD are more sensitive to its effects than offspring exposed to the maternal control diet. Taken together, the results suggest that a maternal HSD and MC-4 receptors play an important role in cocaine relapse.


Subject(s)
Cocaine-Related Disorders/etiology , Cocaine/administration & dosage , Diet/adverse effects , Drug-Seeking Behavior , Extinction, Psychological , Receptor, Melanocortin, Type 4/metabolism , Sugars/toxicity , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/pathology , Female , Male , Maternal Exposure/adverse effects , Pregnancy , Rats, Wistar , Receptor, Melanocortin, Type 4/genetics , Self Administration
19.
Mol Psychiatry ; 25(8): 1759-1776, 2020 08.
Article in English | MEDLINE | ID: mdl-30283033

ABSTRACT

Cocaine-induced vasoconstriction reduces blood flow, which can jeopardize neuronal function and in the prefrontal cortex (PFC) it may contribute to compulsive cocaine intake. Here, we used integrated optical imaging in a rat self-administration and a mouse noncontingent model, to investigate whether changes in the cerebrovascular system in the PFC contribute to cocaine self-administration, and whether they recover with detoxification. In both animal models, cocaine induced severe vasoconstriction and marked reductions in cerebral blood flow (CBF) in the PFC, which were exacerbated with chronic exposure and with escalation of cocaine intake. Though there was a significant proliferation of blood vessels in areas of vasoconstriction (angiogenesis), CBF remained reduced even after 1 month of detoxification. Treatment with Nifedipine (Ca2+ antagonist and vasodilator) prevented cocaine-induced CBF decreases and neuronal Ca2+ changes in the PFC, and decreased cocaine intake and blocked reinstatement of drug seeking. These findings provide support for the hypothesis that cocaine-induced CBF reductions lead to neuronal deficits that contribute to hypofrontality and to compulsive-like cocaine intake in addiction, and document that these deficits persist at least one month after detoxification. Our preliminary data showed that nifedipine might be beneficial in preventing cocaine-induced vascular toxicity and in reducing cocaine intake and preventing relapse.


Subject(s)
Cocaine-Related Disorders/etiology , Cocaine/administration & dosage , Cocaine/pharmacology , Ischemia/chemically induced , Animals , Drug-Seeking Behavior/drug effects , Male , Mice , Nifedipine/pharmacology , Prefrontal Cortex/blood supply , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Self Administration
20.
Neurochem Res ; 46(11): 2909-2922, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34245421

ABSTRACT

Cocaine is a highly addictive stimulant with diverse effects on physiology. Recent studies indicate the involvement of extracellular vesicles (EVs) secreted by neural cells in the cocaine addiction process. It is hypothesized that cocaine affects secretion levels of EVs and their cargos, resulting in modulation of synaptic transmission and plasticity related to addiction physiology and pathology. Lipids present in EVs are important for EV formation and for intercellular lipid exchange that may trigger physiological and pathological responses, including neuroplasticity, neurotoxicity, and neuroinflammation. Specific lipids are highly enriched in EVs compared to parent cells, and recent studies suggest the involvement of various lipids in drug-induced synaptic plasticity during the development and maintenance of addiction processes. Therefore, we examined interstitial small EVs isolated from the brain of mice treated with either saline or cocaine, focusing on the effects of cocaine on the lipid composition of EVs. We demonstrate that 12 days of noncontingent repeated cocaine (10 mg/kg) injections to mice, which induce locomotor sensitization, cause lipid composition changes in brain EVs of male mice as compared with saline-injected controls. The most prominent change is the elevation of GD1a ganglioside in brain EVs of males. However, cocaine does not affect the EV lipid profiles of the brain in female mice. Understanding the relationship between lipid composition in EVs and vulnerability to cocaine addiction may provide insight into novel targets for therapies for addiction.


Subject(s)
Brain/drug effects , Cocaine/toxicity , Dopamine Uptake Inhibitors/toxicity , Extracellular Vesicles/drug effects , Lipid Metabolism/drug effects , Sex Characteristics , Animals , Brain/metabolism , Brain/pathology , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Female , Injections, Intraperitoneal , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL