Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neurosci ; 43(22): 4019-4032, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37094933

ABSTRACT

Dysregulation of the input from the prefrontal cortex (PFC) to the nucleus accumbens (NAc) contributes to cue-induced opioid seeking but the heterogeneity in, and regulation of, prelimbic (PL)-PFC to NAc (PL->NAc) neurons that are altered has not been comprehensively explored. Recently, baseline and opiate withdrawal-induced differences in intrinsic excitability of Drd1+ (D1+) versus Drd2+ (D2+) PFC neurons have been demonstrated. Thus, here we investigated physiological adaptations of PL->NAc D1+ versus D2+ neurons after heroin abstinence and cue-induced relapse. Drd1-Cre+ and Drd2-Cre+ transgenic male Long-Evans rats with virally labeled PL->NAc neurons were trained to self-administer heroin followed by 1 week of forced abstinence. Heroin abstinence significantly increased intrinsic excitability in D1+ and D2+ PL->NAc neurons and increased postsynaptic strength selectively in D1+ neurons. These changes were normalized by cue-induced relapse to heroin seeking. Based on protein kinase A (PKA)-dependent changes in the phosphorylation of plasticity-related proteins in the PL cortex during abstinence and cue-induced relapse to cocaine seeking, we assessed whether the electrophysiological changes in D1+ and D2+ PL->NAc neurons during heroin abstinence were regulated by PKA. In heroin-abstinent PL slices, application of the PKA antagonist (R)-adenosine, cyclic 3',5'-(hydrogenphosphorothioate) triethylammonium (RP-cAMPs) reversed intrinsic excitability in both D1+ and D2+ neurons and postsynaptic strength in only D1+ neurons. Additionally, in vivo bilateral intra-PL infusion of RP-cAMPs after abstinence from heroin inhibited cue-induced relapse to heroin seeking. These data reveal that PKA activity in D1+ and D2+ PL->NAc neurons is not only required for abstinence-induced physiological adaptations but is also required for cue-induced relapse to heroin seeking.SIGNIFICANCE STATEMENT Neuronal plasticity in the medial prefrontal cortex is thought to underlie relapse to drug seeking, yet the subpopulation of neurons that express this plasticity to functionally guide relapse is unclear. Here we show cell type-specific adaptations in Drd1-expressing versus Drd2-expressing prelimbic pyramidal neurons with efferent projections to nucleus accumbens. These adaptations are bidirectionally regulated during abstinence versus relapse and involve protein kinase A (PKA) activation. Furthermore, we show that disruption of the abstinence-associated adaptations via site-specific PKA inhibition abolishes relapse. These data reveal the promising therapeutic potential of PKA inhibition for preventing relapse to heroin seeking and suggest that cell type-specific pharmacologies that target subpopulations of prefrontal neurons would be ideal for future therapeutic developments.


Subject(s)
Cocaine , Nucleus Accumbens , Rats , Animals , Male , Nucleus Accumbens/physiology , Heroin , Rats, Sprague-Dawley , Cyclic AMP-Dependent Protein Kinases/metabolism , Cues , Rats, Long-Evans , Neurons/physiology , Neuronal Plasticity , Recurrence , Receptors, Dopamine D2/metabolism
2.
Addict Biol ; 26(1): e12859, 2021 01.
Article in English | MEDLINE | ID: mdl-31782218

ABSTRACT

Nicotine use remains highly prevalent with tobacco and e-cigarette products consumed worldwide. However, increasing evidence of transgenerational epigenetic inheritance suggests that nicotine use may alter behavior and neurobiology in subsequent generations. We tested the effects of chronic paternal nicotine exposure in C57BL6/J mice on fear conditioning in F1 and F2 offspring, as well as conditioned fear extinction and spontaneous recovery, nicotine self-administration, hippocampal cholinergic functioning, RNA expression, and DNA methylation in F1 offspring. Paternal nicotine exposure was associated with enhanced contextual and cued fear conditioning and spontaneous recovery of extinguished fear memories. Further, nicotine reinforcement was reduced in nicotine-sired mice, as assessed in a self-administration paradigm. These behavioral phenotypes were coupled with altered response to nicotine, upregulated hippocampal nicotinic acetylcholine receptor binding, reduced evoked hippocampal cholinergic currents, and altered methylation and expression of hippocampal genes related to neural development and plasticity. Gene expression analysis suggests multigenerational effects on broader gene networks potentially involved in neuroplasticity and mental disorders. The changes in fear conditioning similarly suggest phenotypes analogous to anxiety disorders similar to post-traumatic stress.


Subject(s)
Fear/drug effects , Hippocampus/metabolism , Memory/drug effects , Nicotine/pharmacology , Paternal Exposure/adverse effects , Animals , Conditioning, Psychological/drug effects , Cues , Extinction, Psychological , Female , Male , Mice , Mice, Inbred C57BL , Up-Regulation/drug effects
3.
Neuropsychopharmacology ; 45(5): 866-876, 2020 04.
Article in English | MEDLINE | ID: mdl-31752015

ABSTRACT

Cognitive flexibility is the ability to switch strategic responses adaptively in changing environments. Cognitive rigidity imposed by neural circuit adaptations during nicotine abstinence may foster maladaptive nicotine taking in addicts. We systematically examined the effects of spontaneous withdrawal in mice exposed to either nicotine (6.3 or 18 mg/kg/day) or saline for 14 days on cognitive flexibility using an operant strategy set-shifting task. Because frontostriatal circuits are critical for cognitive flexibility and brain-derived neurotrophic factor (BDNF) modulates glutamate plasticity in these circuits, we also explored the effects of nicotine withdrawal on these neurochemical substrates. Mice undergoing nicotine withdrawal required more trials to attain strategy-switching criterion. Error analysis show that animals withdrawn from both nicotine doses committed higher perseverative errors, which correlated with measures of anxiety. However, animals treated with the higher nicotine dose also displayed more strategy maintenance errors that remained independent of negative affect. BDNF mRNA expression increased in the medial prefrontal cortex (mPFC) following nicotine withdrawal. Surprisingly, BDNF protein declined in mPFC but was elevated in dorsal striatum (DS). DS BDNF protein positively correlated with perseverative and maintenance errors, suggesting mPFC-DS overflow of BDNF during withdrawal. BDNF-evoked glutamate release and synapsin phosphorylation was attenuated within DS synapses, but enhanced in the nucleus accumbens, suggesting a dichotomous role of BDNF signaling in striatal regions. Taken together, these data suggest that spontaneous nicotine withdrawal impairs distinct components of cognitive set-shifting and these deficits may be linked to BDNF-mediated alterations in glutamate signaling dynamics in discrete frontostriatal circuits.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Frontal Lobe/drug effects , Glutamic Acid/metabolism , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , Adaptation, Physiological/drug effects , Animals , Behavior, Animal , Cognition/drug effects , Cognition/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Corpus Striatum/metabolism , Discrimination, Psychological/drug effects , Discrimination, Psychological/physiology , Frontal Lobe/metabolism , Male , Mice, Inbred C57BL , Synaptic Transmission/drug effects
4.
Sci Adv ; 5(10): eaax7031, 2019 10.
Article in English | MEDLINE | ID: mdl-31633029

ABSTRACT

Smoking is the largest preventable cause of death and disease in the United States. However, <5% of quit attempts are successful, underscoring the urgent need for novel therapeutics. Microglia are one untapped therapeutic target. While previous studies have shown that microglia mediate both inflammatory responses in the brain and brain plasticity, little is known regarding their role in nicotine dependence and withdrawal phenotypes. Here, we examined microglial changes in the striatum-a mesolimbic region implicated in the rewarding effects of drugs and the affective disruptions occurring during withdrawal. We show that both nicotine and withdrawal induce microglial morphological changes; however, proinflammatory effects and anxiogenic behaviors were observed only during nicotine withdrawal. Pharmacological microglial depletion during withdrawal prevented these effects. These results define differential effects of nicotine and withdrawal on inflammatory signaling in the brain, laying the groundwork for development of future smoking cessation therapeutics.


Subject(s)
Microglia/pathology , Nucleus Accumbens/metabolism , Substance Withdrawal Syndrome/pathology , Animals , Anxiety/etiology , Disease Models, Animal , Locomotion , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , NADPH Oxidase 2/metabolism , Nicotine/administration & dosage , Organic Chemicals/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/metabolism
5.
Elife ; 82019 09 05.
Article in English | MEDLINE | ID: mdl-31487241

ABSTRACT

Deficient motivation contributes to numerous psychiatric disorders, including withdrawal from drug use, depression, schizophrenia, and others. Nucleus accumbens (NAc) has been implicated in motivated behavior, but it remains unclear whether motivational drive is linked to discrete neurobiological mechanisms within the NAc. To examine this, we profiled cohorts of Sprague-Dawley rats in a test of motivation to consume sucrose. We found that substantial variability in willingness to exert effort for reward was not associated with operant responding under low-effort conditions or stress levels. Instead, effort-based motivation was mirrored by a divergent NAc shell transcriptome with differential regulation at potassium and dopamine signaling genes. Functionally, motivation was inversely related to excitability of NAc principal neurons. Furthermore, neuronal and behavioral outputs associated with low motivation were linked to faster inactivation of a voltage-gated potassium channel, Kv1.4. These results raise the prospect of targeting Kv1.4 gating in psychiatric conditions associated with motivational dysfunction.


Subject(s)
Kv1.4 Potassium Channel/metabolism , Motivation , Neurons/enzymology , Neurons/physiology , Nucleus Accumbens/physiology , Reward , Animals , Rats, Sprague-Dawley
6.
Behav Brain Res ; 359: 836-844, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30053462

ABSTRACT

Smokers that begin during adolescence are more likely to develop nicotine dependence than those who begin as adults. However, the factors that contribute to this remain largely unknown. Here we utilized a novel operant oral nicotine self-administration procedure in mice to assess the consequences of adolescent nicotine exposure on nicotine and saccharin (non-drug) reinforcement in adults. Animals were given non-contingent exposure to either saline or nicotine using the osmotic minipumps during both adolescence and adulthood for 2 weeks. Reinforcing efficacy for oral nicotine and saccharin was assessed using the progressive ratio schedule 2-weeks following the washout period in adults. Non-contingent nicotine exposure in adolescence drastically increased operant responding for oral nicotine but reduced responding for oral saccharin in the group re-exposed to nicotine in adulthood. Interestingly, adolescent nicotine-exposed mice that received saline exposure as adults exhibited higher preference for oral saccharin. However, breakpoints for oral nicotine in these mice remained comparable to control animals. Surprisingly, both adolescent and adult nicotine exposure increased inactive lever responding during self-administration presumably reflecting impulsive responding. Our data suggest that adolescent nicotine exposure produces an increase in reinforcement sensitivity in adulthood as reflected by increased saccharin self-administration but this sensitivity becomes biased towards nicotine self-administration when re-exposed to nicotine in adulthood. Moreover, nicotine/saccharin reinforcement could be impacted by changes in cognitive control, such as increased impulsivity. These distinct behavioral mechanisms may act in concert to facilitate maladaptive nicotine taking in smokers that initiate nicotine use during adolescence.


Subject(s)
Aging/physiology , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Saccharin/administration & dosage , Sweetening Agents/administration & dosage , Administration, Oral , Aging/drug effects , Animals , Conditioning, Operant/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Reinforcement Schedule , Self Administration , Sex Factors
7.
J Psychopharmacol ; 32(3): 367-372, 2018 03.
Article in English | MEDLINE | ID: mdl-29493350

ABSTRACT

Anxiety and stress disorders have been linked to deficits in fear extinction. Our laboratory and others have demonstrated that acute nicotine impairs contextual fear extinction, suggesting that nicotine exposure may have negative effects on anxiety and stress disorder symptomatology. However, the neurobiological mechanisms underlying the acute nicotine-induced impairment of contextual fear extinction are unknown. Therefore, based on the previous studies showing that brain-derived neurotrophic factor is central for fear extinction learning and acute nicotine dysregulates brain-derived neurotrophic factor signaling, we hypothesized that the nicotine-induced impairment of contextual fear extinction may involve changes in tyrosine receptor kinase B signaling. To test this hypothesis, we systemically, intraperitoneally, injected C57BL/6J mice sub-threshold doses (2.5 and 4.0 mg/kg) of 7,8-dihydroxyflavone, a small-molecule tyrosine receptor kinase B agonist that fully mimics the effects of brain-derived neurotrophic factor, or vehicle an hour before each contextual fear extinction session. Mice also received injections, intraperitoneally, of acute nicotine (0.18 mg/kg) or saline 2-4 min before extinction sessions. While the animals that received only 7,8-dihydroxyflavone did not show any changes in contextual fear extinction, 4.0 mg/kg of 7,8-dihydroxyflavone ameliorated the extinction deficits in mice administered acute nicotine. Overall, these results suggest that acute nicotine-induced impairment of context extinction may be related to a disrupted brain-derived neurotrophic factor signaling.


Subject(s)
Extinction, Psychological/drug effects , Fear/drug effects , Nicotine/pharmacology , Receptors, Amino Acid/agonists , Animals , Anxiety/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Classical/drug effects , Flavones/pharmacology , Mice , Mice, Inbred C57BL , Nicotinic Agonists/pharmacology , Small Molecule Libraries/pharmacology
8.
Neuropsychopharmacology ; 43(6): 1343-1354, 2018 05.
Article in English | MEDLINE | ID: mdl-29114105

ABSTRACT

Neuregulin 3 (NRG3) and ErbB4 have been linked to nicotine addiction; however, the neuronal mechanisms and behavioral consequences of NRG3-ErbB4 sensitivity to nicotine remain elusive. Recent literature suggests that relapse to smoking is due to a lack of impulsive control, which is thought to be due to altered functioning within the orbitofrontal cortex (OFC). Therefore, we examined circuitry changes within this structure following nicotine application. We report that nicotine controls synaptic plasticity in the OFC through NRG3/ErbB4-dependent regulation of GABAergic inhibition. We observed that both nicotine and NRG3 facilitated the conversion of long-term potentiation into long-term depression at cortical layer 3/5 synapses. Induction of long-term depression by nicotine relied on nicotinic receptor activation and key regulators of NRG3 signaling: (1) release of intracellular calcium, (2) activation of the BACE1 beta-secretase, and (3) ErbB4 receptor activation. Nicotine-induced synaptic plasticity was also associated with accumulation of intracellular GABA and was completely blocked by GABAA/GABAB antagonists. To test whether these mechanisms underlie OFC-dependent behavior, we evaluated the effects of nicotine in the go/no-go task. Nicotine-impaired stimulus discrimination in this task was rescued by pharmacologic disruption of the NRG3 receptor, ErbB4. Altogether, our data indicate that nicotine-induced synaptic plasticity in the OFC and cognitive changes depend on NRG3-ErbB4 signaling. We propose that nicotine activation of this pathway may contribute to nicotine addiction, particularly in individuals with genetic variation in NRG3.


Subject(s)
Cognition/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Neuronal Plasticity/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Prefrontal Cortex/drug effects , Animals , Cognition/physiology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice, 129 Strain , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuregulins , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/metabolism , Receptor, ErbB-4/metabolism , Signal Transduction , Synapses/drug effects , Synapses/metabolism , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
9.
J Neurosci Methods ; 277: 30-37, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27939962

ABSTRACT

BACKGROUND: Sustained attention, the ability to detect rare and unpredictable events, is central to cognitive performance. This construct can be tested in rodents using a Sustained Attention Task (SAT), where rats are trained to detect an unpredictably occurring signal (a brief light presentation) from non-signal events. The traditional version of this task utilizes an operant chamber with a central panel light for the signal and two retractable response levers. Adaptation of SAT to the increasingly popular touchscreen operant chambers, which do not have levers or fixed lights, could enhance the versatility of the task. NEW METHOD: Here we developed a touchscreen version of SAT where the light signal is presented in the center of the touchscreen, followed by a tone to indicate the beginning of the response period. Rats indicate their choice during this period by touching their nose to one of two touchscreen response areas. The remaining parameters were kept similar to the traditional version. RESULTS: Rats acquired touchscreen SAT at a similar rate to the traditional version. As with the traditional version, shorter stimulus durations on the signaled trials reduced accuracy and the presence of a distractor (a flashing houselight) disrupted performance on the touchscreen version. COMPARISON TO EXISTING METHOD: Collectively, these data suggest that the touchscreen version is comparable to the traditional version of the SAT, and is an equally valid way of measuring sustained attention. CONCLUSIONS: Many researchers with touchscreen chambers could easily implement our modifications in order to study sustained attention.


Subject(s)
Attention/physiology , Conditioning, Operant/physiology , Psychomotor Performance/physiology , Touch , Animals , Choice Behavior , Male , Rats , Rats, Sprague-Dawley , Reinforcement Schedule , Reward , Statistics, Nonparametric
10.
Neurobiol Learn Mem ; 128: 110-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26775017

ABSTRACT

Nicotine is a major psychoactive and addictive component of tobacco. Although cessation of tobacco use produces various somatic and affective symptoms, withdrawal-related cognitive deficits are considered to be a critical symptom that predict relapse. Therefore, delineating the cognitive mechanisms of nicotine withdrawal may likely provide gainful insights into the neurobiology of nicotine addiction. The present study was designed to examine the effects of nicotine withdrawal induced by mecamylamine, a non-specific nicotinic receptor (nAChR) antagonist, on cognitive control processes in mice using an operant strategy switching task. Brain-derived neurotrophic factor (BDNF) modulates synaptic transmission in frontostriatal circuits, and these circuits are critical for executive functions. Thus, we examined the effects of mecamylamine-precipitated nicotine withdrawal on prefrontal and striatal BDNF protein expression. Mice undergoing precipitated nicotine withdrawal required more trials to attain strategy switching criterion as compared to the controls. Error analysis indicated that impaired performance in these animals was mostly related to their inability to execute the new strategy. The striatal/prefrontal BDNF ratios robustly increased following precipitated nicotine withdrawal. Moreover, higher BDNF ratios were associated with longer task acquisition. Collectively, our findings illustrate that mecamylamine-induced nicotine withdrawal disrupts cognitive control processes and that these changes are possibly linked to perturbations in frontostriatal BDNF signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Executive Function/physiology , Mecamylamine/administration & dosage , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Substance Withdrawal Syndrome/metabolism , Animals , Cognition/drug effects , Cognition/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Executive Function/drug effects , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
11.
Behav Brain Res ; 296: 30-34, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26306826

ABSTRACT

Stressful life events and stress-related psychiatric disorders impair sustained attention, the ability to monitor rare and unpredictable stimulus events over prolonged periods of time. Despite the link between stress and attentional disruptions, the neurobiological basis for stress regulation of attention systems remains underexplored. Here we examined whether corticotropin releasing factor (CRF), which orchestrates stress responses and is hypersecreted in patients with stress-related psychiatric disorders, impairs sustained attention. To this end, male and female rats received central infusions of CRF prior to testing on an operant sustained attention task (SAT), where rats were trained to discriminate signaled from non-signaled events. CRF caused a dose-dependent decrease in SAT performance in both male and female rats. Females were more impaired than males following a moderate dose of CRF, particularly during the middle part of the session. This sex difference was moderated by ovarian hormones. Females in the estrous cycle stage characterized by lower ovarian hormones had a greater CRF-induced attentional impairment than males and females in other cycle stages. Collectively, these studies highlight CRF as a critical stress-related factor that can regulate attentional performance. As sustained attention subserves other cognitive processes, these studies suggest that mitigating high levels of CRF in patients with stress-related psychiatric disorders may ameliorate their cognitive deficits.


Subject(s)
Attention/drug effects , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/pharmacology , Estrous Cycle/metabolism , Psychomotor Performance/drug effects , Animals , Corticotropin-Releasing Hormone/administration & dosage , Dose-Response Relationship, Drug , Female , Male , Rats , Rats, Sprague-Dawley , Sex Factors , Stress, Psychological/metabolism
12.
Psychopharmacology (Berl) ; 232(7): 1207-17, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25281224

ABSTRACT

RATIONALE: Deficits in executive functions underlie compulsive drug use, and understanding how nicotine influences these cognitive processes may provide important information on neurobiological substrates of nicotine addiction. Accumulating evidence suggests that ß2 subunit-containing nicotinic receptors (nAChRs) are involved in the reinforcing process of nicotine addiction. Whether these nAChRs also contributes to the detrimental effects of chronic nicotine on flexible decision-making is not known. OBJECTIVES: In the present study, the effects of chronic nicotine were assessed in mice with partial or complete deletion of the ß2 subunit-containing nAChR gene (ß2+/- or ß2-/-) performing an operant cognitive flexibility task. RESULTS: Visual discrimination learning was not affected in saline-treated ß2 nAChR mutants as compared to the wild-type (ß2+/+) mice; yet, chronic nicotine facilitated acquisition of visual discrimination in all genotypes. The acquisition of new egocentric response strategy set-shifting remained similar in all genotypes, and there was no effect of treatment. Chronic nicotine treatment impaired reversal learning in ß2+/+ mice by increasing response perseveration to the previously rewarded stimulus. Moreover, the acquisition of inverted stimulus-reward contingencies did not differ between ß2+/+ and ß2-/- mice exposed to chronic nicotine. Interestingly, nicotine-induced reversal learning deficits were not observed in ß2+/- mice. CONCLUSIONS: Collectively, these findings suggest that ß2 subunit-containing nAChRs are not critical for visual discrimination learning and extra dimensional rule shift. However, sustained activation of these nAChRs with nicotine may interfere with inhibitory control processes influencing affective shifts in stimulus-reward contingencies.


Subject(s)
Cognition/drug effects , Cognition/physiology , Discrimination Learning/drug effects , Discrimination Learning/physiology , Nicotine/administration & dosage , Receptors, Nicotinic/physiology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Drug Administration Schedule , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Photic Stimulation/methods , Protein Subunits/agonists , Protein Subunits/physiology , Reversal Learning/drug effects , Reversal Learning/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...