Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Addict Biol ; 23(1): 247-255, 2018 01.
Article in English | MEDLINE | ID: mdl-28419646

ABSTRACT

Orexins ('hypocretins') are peptides produced by neurons of the hypothalamus that project to structures implicated in reward and emotion processing. Converging evidence demonstrates functional roles of orexin signaling in arousal, sleep/wakefulness and motivated behaviors for natural and drug rewards. Suvorexant, a dual orexin receptor antagonist, recently received approval from the US Food and Drug Administration to treat insomnia. In Experiment 1, rats self-administered cocaine under a progressive-ratio schedule of reinforcement and the effects of suvorexant on motivation to self-administer cocaine were measured. In Experiment 2, the effects of suvorexant on cocaine reward were assessed by using a place conditioning paradigm, and 50-kHz ultrasonic vocalizations were also recorded to track changes in hedonic reactivity to cocaine. To rule out potentially confounding effects of suvorexant-induced somnolence, locomotor activity was also measured. In Experiment 3, the effects of suvorexant on cocaine-evoked elevations in ventral striatal dopamine were examined. Data reveal that suvorexant (i) reduced the number of cocaine infusions earned during progressive-ratio self-administration; (ii) attenuated initial positive hedonic reactivity to cocaine and prevented cocaine place preference; (iii) did not affect cocaine-induced hyperlocomotion and (iv) reduced cocaine-induced elevations in extracellular ventral striatal dopamine. The present study examined the therapeutic potential of suvorexant in rodent models of cocaine use disorder. These results contribute toward a growing literature supporting therapeutic roles of orexin receptor antagonists in treating substance use disorders.


Subject(s)
Azepines/pharmacology , Behavior, Animal/drug effects , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Motivation/drug effects , Orexin Receptor Antagonists/pharmacology , Triazoles/pharmacology , Ventral Striatum/drug effects , Animals , Cocaine/pharmacology , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Locomotion/drug effects , Male , Rats , Reinforcement, Psychology , Reward , Self Administration , Ventral Striatum/metabolism
2.
Addict Biol ; 23(1): 102-110, 2018 01.
Article in English | MEDLINE | ID: mdl-27910188

ABSTRACT

Abuse of synthetic psychostimulants like synthetic cathinones has risen in recent years. 3,4-Methylenedioxypyrovalerone (MDPV) is one such synthetic cathinone that demonstrates a mechanism of action similar to cocaine. Compared to cocaine, MDPV is more potent at blocking dopamine and norepinephrine reuptake and is readily self-administered by rodents. The present study compared the rewarding and reinforcing properties of MDPV and cocaine using systemic injection dose-response and self-administration models. Fifty kilohertz ultrasonic vocalizations (USVs) were recorded as an index of positive affect throughout experiments. In Experiment 1, MDPV and cocaine dose-dependently elicited 50-kHz USVs upon systemic injection, but MDPV increased USVs at greater rates and with greater persistence relative to cocaine. In Experiment 2, latency to begin MDPV self-administration was shorter than latency to begin cocaine self-administration, and self-administered MDPV elicited greater and more persistent rates of 50-kHz USVs versus cocaine. MDPV-elicited 50-kHz USVs were sustained over the course of drug load-up whereas cocaine-elicited USVs waned following initial infusions. Notably, we observed a robust presence of context-elicited 50-kHz USVs from both MDPV and cocaine self-administering rats. Collectively, these data suggest that MDPV has powerfully rewarding and reinforcing effects relative to cocaine at one-tenth doses. Consistent with prior work, we additionally interpret these data in supporting that MDPV has significant abuse risk based on its potency and subjectively positive effects. Future studies will be needed to better refine therapeutic strategies targeted at reducing the rewarding effects of cathinone analogs in efforts to ultimately reduce abuse liability.


Subject(s)
Benzodioxoles/pharmacology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Pyrrolidines/pharmacology , Reward , Vocalization, Animal/drug effects , Animals , Benzodioxoles/administration & dosage , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Male , Pyrrolidines/administration & dosage , Rats , Reinforcement, Psychology , Self Administration , Ultrasonic Waves , Synthetic Cathinone
3.
Behav Pharmacol ; 28(6): 485-488, 2017 09.
Article in English | MEDLINE | ID: mdl-28704277

ABSTRACT

Nicotine is the addictive compound in tobacco products which exerts psychosomatic effects that contribute to abuse and to low rates of abstinence in treatment-seeking smokers. At present, the most successful smoking cessation aide helps one in four individuals quit smoking at 1 year postcessation. New adjunctive therapies are needed to improve status of smoking-related public health crises, and ß-lactam antibiotics are one class of potential therapies as they favorably augment extrasynaptic glutamate clearance. Our study used two-chamber place conditioning to assess effects of ceftriaxone (CTX) on persistence of conditioned nicotine reward. Rats were conditioned to associate nicotine (0.4 mg/kg, subcutaneous) with one context and vehicle with an alternative context. After initial post-test, rats received either daily ceftriaxone (200 mg/kg, intraperitoneal) or saline. All rats showed nicotine place preference during post-test 1. CTX-treated rats meeting extinction criterion by post-test 7 showed significantly reduced preference for the nicotine-paired context during post-test 2 compared with vehicle-treated rats. We interpret these data to support the further study of CTX as a smoking cessation aide. Our results suggest that CTX reduces persistence of conditioned nicotine reward and may be helpful for improving abstinence rates in a subset of treatment-seeking smokers.


Subject(s)
Ceftriaxone/pharmacology , Conditioning, Classical/drug effects , Nicotine/pharmacology , Animals , Conditioning, Psychological/drug effects , Male , Nicotine/metabolism , Nicotinic Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Reward , Smoking , Smoking Cessation/methods , Tobacco Use Disorder/psychology
4.
Proc Natl Acad Sci U S A ; 111(16): E1648-55, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24706819

ABSTRACT

Hypocretin (orexin) and dynorphin are neuropeptides with opposing actions on motivated behavior. Orexin is implicated in states of arousal and reward, whereas dynorphin is implicated in depressive-like states. We show that, despite their opposing actions, these peptides are packaged in the same synaptic vesicles within the hypothalamus. Disruption of orexin function blunts the rewarding effects of lateral hypothalamic (LH) stimulation, eliminates cocaine-induced impulsivity, and reduces cocaine self-administration. Concomitant disruption of dynorphin function reverses these behavioral changes. We also show that orexin and dynorphin have opposing actions on excitability of ventral tegmental area (VTA) dopamine neurons, a prominent target of orexin-containing neurons, and that intra-VTA orexin antagonism causes decreases in cocaine self-administration and LH self-stimulation that are reversed by dynorphin antagonism. Our findings identify a unique cellular process by which orexin can occlude the reward threshold-elevating effects of coreleased dynorphin and thereby act in a permissive fashion to facilitate reward.


Subject(s)
Dynorphins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Reward , Ventral Tegmental Area/metabolism , Animals , Cocaine/administration & dosage , Cocaine/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dynorphins/antagonists & inhibitors , Hypothalamus/drug effects , Hypothalamus/metabolism , Impulsive Behavior/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/antagonists & inhibitors , Orexin Receptors/metabolism , Orexins , Self Administration , Synaptic Transmission/drug effects , Ventral Tegmental Area/drug effects
5.
J Neurosci ; 31(8): 3095-103, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21414930

ABSTRACT

Stress triggers psychiatric conditions including depressive and anxiety disorders. The mechanisms by which stress produces persistent changes in behavior are not fully understood. Here we show in rats that stress (footshock) activates the transcription factor cAMP response element binding protein (CREB) within the nucleus accumbens shell (NAS), a brain area involved in encoding reward and aversion. To examine the behavioral significance of altered CREB function in the NAS, we used viral vectors to elevate or disrupt CREB in this region. Elevated CREB produced increases in intracranial self-stimulation thresholds, a depressive-like sign reflecting anhedonia (decreased sensitivity to reward), whereas disruption of CREB function by expression of a dominant-negative CREB had the opposite effect. To determine whether neuroadaptations that produce anhedonia subsequently affect vulnerability to stress-induced behavioral adaptations, we subjected rats with altered CREB function in the NAS to fear conditioning. Although neither elevation nor disruption of CREB function altered the development of conditioned fear, elevation of CREB impaired extinction of conditioned fear. To mimic downstream effects of CREB activation on expression of the opioid peptide dynorphin, we microinjected the κ-opioid receptor (KOR) agonist U50,488 directly into the NAS. KOR stimulation produced anhedonia but had no effect on expression or extinction of conditioned fear. These findings demonstrate that activation of CREB in the NAS produces multiple behavioral signs (anhedonia, impaired extinction) characteristic of experience-dependent psychiatric conditions such as posttraumatic stress disorder. Although CREB activation is a common trigger, expression of these individual signs appears to involve divergent downstream mechanisms.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Extinction, Psychological/physiology , Fear/physiology , Nucleus Accumbens/metabolism , Reward , Stress, Psychological/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/genetics , Stress, Psychological/physiopathology
6.
Neuropsychopharmacology ; 46(12): 2197-2206, 2021 11.
Article in English | MEDLINE | ID: mdl-34408277

ABSTRACT

Brain α2-containing GABAA receptors play a critical role in the modulation of anxiety- and fear-like behavior. However, it is unknown whether these receptors also play a role in modulating resilience to chronic stress, and in which brain areas and cell types such an effect would be mediated. We evaluated the role of α2-containing GABAA receptors following chronic social defeat stress using male mice deficient in the α2 subunit globally or conditionally in dopamine D1- or D2-receptor-expressing neurons, e.g., within the nucleus accumbens (NAc). In addition, we examined the effect of the lack of the α2 subunit on intermediates of the glutathione synthesis pathway. We found that α2-containing GABAA receptors on D2-receptor-positive but not on D1-receptor-positive neurons promote resiliency to chronic social defeat stress, as reflected in social interaction tests. The pro-resiliency effects of α2-containing GABAA receptors on D2-receptor-positive neurons do not appear to be directly related to alterations in anxiety-like behavior, as reflected in the elevated plus-maze, light-dark box, and novel open field tests. Increases in indices of oxidative stress-reflected by increases in cystathionine levels and reductions in GSH/GSSG ratios-were found in the NAc and prefrontal cortex but not in the hippocampus of mice lacking α2-containing GABAA receptors. We conclude that α2-containing GABAA receptors within specific brain areas and cell populations promote stress resiliency independently of direct effects on anxiety-like behaviors. A potential mechanism contributing to this increased resiliency is the protection that α2-containing GABAA receptors provide against oxidative stress in NAc and the prefrontal cortex.


Subject(s)
Anxiety , Receptors, GABA-A/metabolism , Receptors, GABA , Animals , Male , Mice , Mice, Inbred C57BL , Receptors, Dopamine D1/metabolism , gamma-Aminobutyric Acid
7.
BMC Res Notes ; 13(1): 384, 2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32799930

ABSTRACT

OBJECTIVE: Selective kappa opioid receptor (KOR) agonists were shown to produce a dose-dependent depression of brain-stimulation reward (BSR) in the rat intracranial self-stimulation (ICSS) tests. However, limited studies using mice produced less conclusive results. Here the effects of U50,488H were re-examined on BSR in mice with a larger cohort of animals. RESULTS: Forty C57BL/6J male mice were implanted with the electrodes in medial forebrain bundle. About a week after surgery, mice were subject to ICSS training. Only eighteen passed the two-phase procedures, at which point they readily spun the wheels to obtain reinforcing effect of BSR, and were used for the ICSS tests. Compared with saline (s.c.), U50,488H (2 mg/kg, s.c.) did not have effects on the BSR thresholds within 1 h post-treatment, while it decreased the maximum wheel-spinning rates in a time-dependent manner. In contrast, cocaine (5 mg/kg, s.c.) decreased the BSR thresholds time-dependently without affecting the maximum wheel-spinning rates in the same cohort of mice, demonstrating the validity of our mouse ICSS models. For comparison, U50,488H (2 mg/kg, s.c.) induced significant conditioned place aversion (CPA) in a different cohort of mice without surgeries. Thus, ICSS may not be an appropriate test for KOR agonist-induced aversion in mice.


Subject(s)
Receptors, Opioid, kappa , Reward , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Receptors, Opioid, kappa/metabolism
8.
Neuropsychopharmacology ; 44(5): 939-949, 2019 04.
Article in English | MEDLINE | ID: mdl-30082888

ABSTRACT

Kappa opioid receptor (KOR) agonists produce analgesic and anti-pruritic effects, but their clinical application was limited by dysphoria and hallucinations. Nalfurafine, a clinically used KOR agonist, does not cause dysphoria or hallucinations at therapeutic doses in humans. We found that in CD-1 mice nalfurafine produced analgesic and anti-scratch effects dose-dependently, like the prototypic KOR agonist U50,488H. In contrast, unlike U50,488H, nalfurafine caused no aversion, anhedonia, or sedation or and a low level of motor incoordination at the effective analgesia and anti-scratch doses. Thus, we established a mouse model that recapitulated important aspects of the clinical observations. We then employed a phosphoproteomics approach to investigate mechanisms underlying differential KOR-mediated effects. A large-scale mass spectrometry (MS)-based analysis on brains revealed that nalfurafine perturbed phosphoproteomes differently from U50,488H in a brain-region specific manner after 30-min treatment. In particular, U50,488H and nalfurafine imparted phosphorylation changes to proteins found in different cellular components or signaling pathways in different brain regions. Notably, we observed that U50,488H, but not nalfurafine, activated the mammalian target of rapamycin (mTOR) pathway in the striatum and cortex. Inhibition of the mTOR pathway by rapamycin abolished U50,488H-induced aversion, without affecting analgesic, anti-scratch, and sedative effects and motor incoordination. The results indicate that the mTOR pathway is involved in KOR agonist-induced aversion. This is the first demonstration that phosphoproteomics can be applied to agonist-specific signaling of G protein-coupled receptors (GPCRs) in mouse brains to unravel pharmacologically important pathways. Furthermore, this is one of the first two reports that the mTOR pathway mediates aversion caused by KOR activation.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Antipruritics/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Morphinans/pharmacology , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects , Spiro Compounds/pharmacology , TOR Serine-Threonine Kinases/drug effects , Animals , Disease Models, Animal , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociception/drug effects , Phosphorylation/drug effects , Proteomics
9.
J Neurosci ; 27(11): 2837-45, 2007 Mar 14.
Article in English | MEDLINE | ID: mdl-17360905

ABSTRACT

The role of hypocretin (orexin; hcrt/orx) neurons in regulation of arousal is well established. Recently, hcrt/orx has been implicated in food reward and drug-seeking behavior. We report here that in male rats, Fos immunoreactivity (ir) in hcrt/orx neurons increases markedly during copulation, whereas castration produces decreases in hcrt/orx neuron cell counts and protein levels in a time course consistent with postcastration impairments in copulatory behavior. This effect was reversed by estradiol replacement. Immunolabeling for androgen (AR) and estrogen (ER alpha) receptors revealed no colocalization of hcrt/orx with AR and few hcrt/orx neurons expressing ER alpha, suggesting that hormonal regulation of hcrt/orx expression is via afferents from neurons containing those receptors. We also demonstrate that systemic administration of the orexin-1 receptor antagonist SB 334867 [N-(2-methyl-6-benzoxazolyl)-N''-1,5-naphthyridin-4-yl urea] impairs copulatory behavior. One locus for the prosexual effects of hcrt/orx may be the ventral tegmental area (VTA). We show here that hcrt-1/orx-A produces dose-dependent increases in firing rate and population activity of VTA dopamine (DA) neurons in vivo. Activation of hcrt/orx during copulation, and in turn, excitation of VTA DA neurons by hcrt/orx, may contribute to the robust increases in nucleus accumbens DA previously observed during male sexual behavior. Subsequent triple immunolabeling in anterior VTA showed that Fos-ir in tyrosine hydroxylase-positive neurons apposed to hcrt/orx fibers increases during copulation. Together, these data support the view that hcrt/orx peptides may act in a steroid-sensitive manner to facilitate the energized pursuit of natural rewards like sex via activation of the mesolimbic DA system.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Sexual Behavior, Animal/physiology , Animals , Female , Gonadal Steroid Hormones/analysis , Gonadal Steroid Hormones/physiology , Intracellular Signaling Peptides and Proteins/analysis , Male , Neurons/chemistry , Neurons/physiology , Neuropeptides/analysis , Orchiectomy , Orexins , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Ventral Tegmental Area/chemistry , Ventral Tegmental Area/physiology
10.
Neuropsychopharmacology ; 43(5): 1001-1009, 2018 04.
Article in English | MEDLINE | ID: mdl-28741623

ABSTRACT

Hypothalamic hypocretin (orexin) peptides mediate arousal, attention, and reward processing. Fibers containing orexins project to brain structures that govern motivated behavior, including the ventral tegmental area (VTA). A number of psychiatric conditions, including attention deficit hyperactivity disorder (ADHD) and substance use disorders, are characterized by deficits in impulse control, however the relationship between orexin and impulsive behavior is incompletely characterized. The effects of systemic or centrally administered orexin receptor (OXR) antagonists on measures of impulsive-like behavior in rats were evaluated using the five-choice serial reaction time task (5-CSRTT) and delay discounting procedures. These paradigms were also used to test the capacity of OXR antagonists to attenuate acute cocaine-evoked impulsivity. Finally, immunohistochemistry and calcium imaging were used to assess potential cellular mechanisms by which OXR blockade may influence motor impulsivity. Suvorexant, a dual (OX1/2R) orexin receptor antagonist, reduced cocaine-evoked premature responses in 5-CSRTT when administered systemically or directly into VTA. Neither suvorexant nor OX1R- or OX2R-selective compounds (SB334867 or TCS-OX2-29, respectively) altered delay discounting. Finally, suvorexant did not alter Fos-immunoreactivity within tyrosine hydroxylase-immunolabeled neurons of VTA, but did attenuate cocaine- and orexin-induced increases in calcium transient amplitude within neurons of VTA. Results from the present studies suggest potential therapeutic utility of OXR antagonists in reducing psychostimulant-induced motor impulsivity. These findings also support the view that orexin transmission is closely involved in executive function in normal and pathological conditions.


Subject(s)
Azepines/pharmacology , Cocaine/antagonists & inhibitors , Cocaine/pharmacology , Impulsive Behavior/drug effects , Orexin Receptor Antagonists/pharmacology , Triazoles/pharmacology , Animals , Benzoxazoles/pharmacology , Choice Behavior , Delay Discounting/drug effects , Isoquinolines/pharmacology , Male , Microinjections , Naphthyridines , Orexins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyridines/pharmacology , Rats , Urea/analogs & derivatives , Urea/pharmacology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
11.
ACS Chem Neurosci ; 9(10): 2379-2394, 2018 10 17.
Article in English | MEDLINE | ID: mdl-29714473

ABSTRACT

Cathinone is a plant alkaloid found in khat leaves of perennial shrubs grown in East Africa. Similar to cocaine, cathinone elicits psychostimulant effects which are in part attributed to its amphetamine-like structure. Around 2010, home laboratories began altering the parent structure of cathinone to synthesize derivatives with mechanisms of action, potencies, and pharmacokinetics permitting high abuse potential and toxicity. These "synthetic cathinones" include 4-methylmethcathinone (mephedrone), 3,4-methylenedioxypyrovalerone (MDPV), and the empathogenic agent 3,4-methylenedioxymethcathinone (methylone) which collectively gained international popularity following aggressive online marketing as well as availability in various retail outlets. Case reports made clear the health risks associated with these agents and, in 2012, the Drug Enforcement Agency of the United States placed a series of synthetic cathinones on Schedule I under emergency order. Mechanistically, cathinone and synthetic derivatives work by augmenting monoamine transmission through release facilitation and/or presynaptic transport inhibition. Animal studies confirm the rewarding and reinforcing properties of synthetic cathinones by utilizing self-administration, place conditioning, and intracranial self-stimulation assays and additionally show persistent neuropathological features which demonstrate a clear need to better understand this class of drugs. This Review will thus detail (i) historical context of cathinone use and the rise of "dark" synthetic derivatives, (ii) structural features and mechanisms of synthetic cathinones, (iii) behavioral effects observed clinically and in animals under controlled laboratory conditions, and (iv) neurotransmitters and circuits that may be targeted to manage synthetic cathinone abuse in humans.


Subject(s)
Alkaloids/chemistry , Alkaloids/pharmacology , Central Nervous System Stimulants/chemistry , Central Nervous System Stimulants/pharmacology , Alkaloids/history , Animals , Behavior, Animal/drug effects , Benzodioxoles/chemistry , Benzodioxoles/history , Benzodioxoles/pharmacology , Body Temperature/drug effects , Catha/chemistry , Central Nervous System Stimulants/history , Dopamine/metabolism , History, 21st Century , History, Medieval , Humans , Locomotion/drug effects , Methamphetamine/analogs & derivatives , Methamphetamine/chemistry , Methamphetamine/history , Methamphetamine/pharmacology , Pyrrolidines/chemistry , Pyrrolidines/history , Pyrrolidines/pharmacology , Serotonin/metabolism , Substance-Related Disorders , Synaptic Transmission/drug effects , Synthetic Cathinone
12.
Neurosci Lett ; 427(3): 123-6, 2007 Nov 12.
Article in English | MEDLINE | ID: mdl-17933463

ABSTRACT

In male rats, estradiol (E(2)) exerts marked anorectic effects. One mechanism proposed for this effect is an E(2)-mediated down-regulation of the orexigenic neuropeptide melanin concentrating hormone (MCH). Previous anatomical work has shown that both MCH and estrogen receptor alpha (ERalpha) are found in quantity in the lateral hypothalamic area (LHA), a structure long associated with appetite and ingestive behavior. It has been hypothesized that the most direct manner by which E(2) could affect MCH expression and feeding would be via classical nuclear ERalpha located in MCH neurons. To evaluate this notion, we performed double-label immunohistochemistry for MCH and ERalpha in male rat hypothalamus. We report here that MCH neurons do not contain ERalpha, suggesting that the primary locus for estrogenic control of feeding is not the MCH neurons themselves. Rather, we show substantial overlap in the anatomical distribution of both cell types, raising the possibility that E(2) influences MCH signaling indirectly via adjacent ERalpha-containing cells.


Subject(s)
Estrogen Receptor alpha/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Animals , Hypothalamus/cytology , Male , Neurons/classification , Rats
13.
Psychopharmacology (Berl) ; 234(21): 3207-3215, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28786030

ABSTRACT

RATIONALE: Synthetic psychostimulant abuse, including cathinone-derived 3,4-methylenedioxypyrovalerone (MDPV), continues to increase in many countries. Similar to cocaine but with greater potency, MDPV elicits a transient sympathomimetic response by blocking cellular uptake of dopamine (DA) and norepinephrine (NE)-administration in some users is reported as euphoria-inducing much like cocaine and amphetamine. Pharmacological agents that disrupt excitatory transmission onto midbrain DA-producing neurons, including hypothalamic hypocretin/orexin (hcrt/ox) receptor antagonists, present attractive targets to aide abstinence maintenance by reducing psychostimulant-associated reward and reinforcement. OBJECTIVE: The present study sought to assess the degree to which suvorexant, a dual hcrt/ox receptor antagonist, influences drug-taking as well as ultrasonic vocalizations (USVs) associated with MDPV self-administration. METHODS: Rats were trained to self-administer MDPV (~0.03 mg/kg/inf, 3-s) for 14 days under a fixed-ratio 1 schedule of reinforcement, and effects of suvorexant (0, 3, 10, 30 mg/kg, i.p.) on drug-taking was assessed. USVs were recorded during a 30-min pre-lever period as well as during 2-h of MDPV self-administration. RESULTS: We observed that suvorexant modestly suppressed the number of MDPV infusions earned. Notably, we observed that suvorexant reduced 50-kHz USVs associated with pre- and post-lever time-points but did not noticeably alter call type profiles. Upon comparison of the two measures, we observed trending positive associations between suvorexant-induced changes in drug-taking and 50-kHz USVs. CONCLUSIONS: Results from this exploratory study provide support for the following: (1) studying how suvorexant may provide benefit to humans with stimulant use disorders, (2) identifying a potential role for orexin transmission in cathinone abuse, and (3) further interrogating the potential utility of rat USVs to predict drug consumption in preclinical models of substance use disorders.


Subject(s)
Azepines/pharmacology , Benzodioxoles/administration & dosage , Craving/drug effects , Dopaminergic Neurons/drug effects , Mesencephalon/drug effects , Orexin Receptors/drug effects , Pyrrolidines/administration & dosage , Triazoles/pharmacology , Vocalization, Animal/drug effects , Animals , Dose-Response Relationship, Drug , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley , Self Administration , Synthetic Cathinone
14.
ACS Chem Neurosci ; 8(12): 2648-2654, 2017 12 20.
Article in English | MEDLINE | ID: mdl-28885007

ABSTRACT

Mephedrone (4-methylmethcathinone (4-MMC)) (MEPH) is a new psychoactive substance (NPS) of the synthetic cathinone class. MEPH has a chiral center and exists as two enantiomers (R-,S-MEPH), yet stereospecific effects of MEPH have not been extensively investigated in preclinical assays. Because significant behavioral and neurochemical differences can exist between enantiomers, probing effects of stereochemistry on biological activity enables separation of adverse and therapeutic effects. Our prior work showed that R-MEPH, relative to S-MEPH, produced greater locomotor activation, place preference, and facilitation of brain reward thresholds in rodents. The present study sought to determine if MEPH enantiomers display stereospecific reward and reinforcement in rat self-administration assays. In Experiment 1, rats were trained to self-administer racemic MEPH (0.50 mg/kg/inf), and dose substitution effects of R-MEPH (0.50 mg/kg/inf) and S-MEPH (0.25, 0.50, 2.00 mg/kg/inf) were examined. In Experiment 2, separate rats were trained to self-administer R-MEPH (0.25, 0.50, 2.00 mg/kg/inf) or S-MEPH (0.25, 0.50, 2.00 mg/kg/inf) and were thereafter evaluated under progressive-ratio access conditions. Within this cohort, 50 kHz ultrasonic vocalizations (USVs) were recorded to measure potential differences in subjective positive affect associated with MEPH enantiomer self-administration. We identified enantiomer- and dose-dependent effects on infusions earned during self-administration following acquisition of racemic MEPH, with greatest infusions under low-effort, fixed-ratio 1 access conditions from low-dose S-MEPH self-administration. When taxed with progressive-ratio access conditions, rats trained to self-administer R-MEPH showed higher break points than those of rats trained to self-administer S-MEPH. Additionally, R-MEPH elicited greatest rates of 50 kHz USVs compared to S-MEPH. Taken together, these data suggest that the R-enantiomer of MEPH is primarily responsible for the rewarding, reinforcing, and motivational properties of racemic MEPH, which increases our understanding of stereospecific preferences pertaining to MEPH abuse.


Subject(s)
Methamphetamine/analogs & derivatives , Motivation/drug effects , Motivation/physiology , Reward , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Dose-Response Relationship, Drug , Male , Methamphetamine/administration & dosage , Methamphetamine/chemistry , Rats , Rats, Sprague-Dawley , Self Administration , Stereoisomerism , Structure-Activity Relationship , Treatment Outcome
15.
Behav Brain Res ; 314: 226-33, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27491589

ABSTRACT

Tobacco smoking is the leading cause of preventable death in the United States. Nicotine is the principal psychoactive ingredient in tobacco that causes addiction. The structures governing nicotine addiction, including those underlying withdrawal, are still being explored. Nicotine withdrawal is characterized by negative affective and cognitive symptoms that enhance relapse susceptibility, and suppressed dopaminergic transmission from ventral tegmental area (VTA) to target structures underlies behavioral symptoms of nicotine withdrawal. Agonist and partial agonist therapies help 1 in 4 treatment-seeking smokers at one-year post-cessation, and new targets are needed to more effectively aid smokers attempting to quit. Hypothalamic orexin/hypocretin neurons send excitatory projections to dopamine (DA)-producing neurons of VTA and modulate mesoaccumbal DA release. The effects of nicotinic receptor blockade, which is commonly used to precipitate withdrawal, on orexin neurons remain poorly investigated and present an attractive target for intervention. The present study sought to investigate the effects of nicotinic receptor blockade on hypothalamic orexin neurons using mecamylamine to precipitate withdrawal in rats. Separate groups of rats were treated with either chronic nicotine or saline for 7-days at which point effects of mecamylamine or saline on somatic signs and anxiety-like behavior were assessed. Finally, tissue from rats was harvested for immunofluorescent analysis of Fos within orexin neurons. Results demonstrate that nicotinic receptor blockade leads to reduced orexin cell activity, as indicated by lowered Fos-immunoreactivity, and suggest that this underlying cellular activity may be associated with symptoms of nicotine withdrawal as effects were most prominently observed in rats given chronic nicotine. We conclude from this study that orexin transmission becomes suppressed in rats upon nicotinic receptor blockade, and that behavioral symptoms associated with nicotine withdrawal may be aided by intervention upon orexinergic transmission.


Subject(s)
Neurons/drug effects , Nicotinic Antagonists/pharmacology , Orexins/metabolism , Receptors, Nicotinic/drug effects , Animals , Dopamine/pharmacology , Immunohistochemistry/methods , Male , Neurons/metabolism , Nicotine/pharmacology , Oncogene Proteins v-fos/metabolism , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
16.
Br J Pharmacol ; 172(2): 334-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24641197

ABSTRACT

UNLABELLED: Addiction is a devastating disorder that affects 15.3 million people worldwide. While prevalent, few effective treatments exist. Orexin receptors have been proposed as a potential target for anti-craving medications. Orexins, also known as hypocretins, are neuropeptides produced in neurons of the lateral and dorsomedial hypothalamus and perifornical area, which project widely throughout the brain. The absence of orexins in rodents and humans leads to narcolepsy. However, orexins also have an established role in reward seeking. This review will discuss some of the original studies describing the roles of the orexins in reward seeking as well as specific works that were presented at the 2013 International Narcotics Research Conference. Orexin signalling can promote drug-induced plasticity of glutamatergic synapses onto dopamine neurons of the ventral tegmental area (VTA), a brain region implicated in motivated behaviour. Additional evidence suggests that orexin signalling can also promote drug seeking by initiating an endocannabinoid-mediated synaptic depression of GABAergic inputs to the VTA, and thereby disinhibiting dopaminergic neurons. Orexin neurons co-express the inhibitory opioid peptide dynorphin. It has been proposed that orexin in the VTA may not mediate reward per se, but rather occludes the 'anti-reward' effects of dynorphin. Finally, orexin signalling in the prefrontal cortex and the central amygdala is implicated in reinstatement of reward seeking. This review will highlight recent work describing the role of orexin signalling in cellular processes underlying addiction-related behaviours and propose novel hypotheses for the mechanisms by which orexin signalling may impart drug seeking. LINKED ARTICLES: This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Reward , Substance-Related Disorders/metabolism , Animals , Dynorphins/metabolism , Ethanol/administration & dosage , Humans , Hypothalamus/metabolism , Orexin Receptors/metabolism , Orexins , Ventral Tegmental Area/metabolism
17.
Behav Neurosci ; 118(6): 1317-23, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15598140

ABSTRACT

Dopamine in the medial preoptic area (MPOA) facilitates copulation in male rats, and nitric oxide (NO) regulates basal and female-stimulated MPOA dopamine release. Microinjection of L-nitro-arginine methyl ester (L-NAME, an NO synthesis inhibitor) into the MPOA blocked copulation in naive rats and impaired copulation in sexually experienced males. In other naive rats, L-NAME or saline was microinjected into the MPOA before each of 7 daily exposures to a receptive female placed over their cage. In a drug-free test on Day 8, copulation by L-NAME-treated rats was similar to that of unexposed controls and was impaired relative to saline-treated males. Therefore, NO in the MPOA is important for copulation and stimulus sensitization in male rats.


Subject(s)
Copulation/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Preoptic Area/drug effects , Analysis of Variance , Animals , Behavior, Animal , Ejaculation/drug effects , Enzyme Inhibitors/pharmacology , Male , Preoptic Area/anatomy & histology , Rats , Rats, Long-Evans , Reaction Time/drug effects
18.
Brain Res ; 1023(1): 134-40, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15364028

ABSTRACT

The ability of hallucinogens to increase extracellular glutamate in the prefrontal cortex (PFC) was assessed by in vivo microdialysis. The hallucinogen lysergic acid diethylamide (LSD; 0.1 mg/kg, i.p.) caused a time-dependent increase in PFC glutamate that was blocked by the 5-HT(2A) antagonist M100907 (0.05 mg/kg, i.p.). Similarly, the 5-HT(2A/C) agonist [-]-2,5-dimethoxy-4-methylamphetamine (DOM; 0.6 mg/kg, i.p.), which is a phenethylamine hallucinogen, increased glutamate to 206% above saline-treated controls. When LSD (10 microM) was directly applied to the PFC by reverse dialysis, a rapid increase in PFC glutamate levels was observed. Glutamate levels in the PFC remained elevated after the drug infusion was discontinued. These data provide direct evidence in vivo for the hypothesis that an enhanced release of glutamate is a common mechanism in the action of hallucinogens.


Subject(s)
DOM 2,5-Dimethoxy-4-Methylamphetamine/pharmacology , Extracellular Fluid/drug effects , Glutamic Acid/metabolism , Lysergic Acid Diethylamide/pharmacology , Prefrontal Cortex/drug effects , Animals , Extracellular Fluid/metabolism , Glutamic Acid/biosynthesis , Male , Prefrontal Cortex/metabolism , Rats , Rats, Inbred F344 , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists , Serotonin 5-HT2 Receptor Antagonists
19.
Physiol Behav ; 83(2): 291-307, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15488546

ABSTRACT

Steroid hormones regulate sexual behavior primarily by slow, genomically mediated effects. These effects are realized, in part, by enhancing the processing of relevant sensory stimuli, altering the synthesis, release, and/or receptors for neurotransmitters in integrative areas, and increasing the responsiveness of appropriate motor outputs. Dopamine has facilitative effects on sexual motivation, copulatory proficiency, and genital reflexes. Dopamine in the nigrostriatal tract influences motor activity; in the mesolimbic tract it activates numerous motivated behaviors, including copulation; in the medial preoptic area (MPOA) it controls genital reflexes, copulatory patterns, and specifically sexual motivation. Testosterone increases nitric oxide synthase in the MPOA; nitric oxide increases basal and female-stimulated dopamine release, which in turn facilitates copulation and genital reflexes. Serotonin (5-HT) is primarily inhibitory, although stimulation of 5-HT(2C) receptors increases erections and inhibits ejaculation, whereas stimulation of 5-HT(1A) receptors has the opposite effects: facilitation of ejaculation and, in some circumstances, inhibition of erection. 5-HT is released in the anterior lateral hypothalamus at the time of ejaculation. Microinjections of selective serotonin reuptake inhibitors there delay the onset of copulation and delay ejaculation after copulation begins. One means for this inhibition is a decrease in dopamine release in the mesolimbic tract.


Subject(s)
Dopamine/physiology , Preoptic Area/physiology , Serotonin/physiology , Sexual Behavior, Animal/physiology , Sexual Behavior/physiology , Animals , Humans , Hypothalamus/physiology , Male , Nitric Oxide/metabolism , Rats , Sex Characteristics , Sex Factors , Testosterone/metabolism
20.
Pharmacol Biochem Behav ; 73(4): 835-42, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12213529

ABSTRACT

The extent to which acute and repeated administration of the CB(1) agonist WIN 55,212-2 would affect the stimulatory properties of amphetamine was assessed in Lewis rats. In the first experiment, Lewis rats were treated with either 1 mg/kg of WIN 55,212-2 or vehicle and subsequently treated with 2 mg/kg amphetamine. Acute treatment with WIN 55,212-2 initially increased locomotor activity and then attenuated the stimulating effect of amphetamine on locomotion and exploration (as measured by rears). In a separate experiment, Lewis rats were given daily injections of either WIN 55,212-2 (1 mg/kg) or vehicle for 10 days and the effects of amphetamine were assessed at 1 and 3 days following the last chronic cannabinoid treatment. Those rats, which had been treated with WIN 55,212-2, had an enhanced response to amphetamine with rearing but not with ambulatory movements, suggesting the occurrence of behavioral cross-sensitization to the ability of amphetamine to increase rearing. These data add to the growing evidence that there is at least some overlap between those neural systems acted upon by cannabinoids and those that are believed to be involved in incentive properties associated with other drugs of abuse.


Subject(s)
Amphetamine/pharmacology , Cannabinoids/administration & dosage , Morpholines/administration & dosage , Motor Activity/drug effects , Naphthalenes/administration & dosage , Receptors, Drug/agonists , Animals , Benzoxazines , Drug Synergism , Male , Motor Activity/physiology , Rats , Rats, Inbred Lew , Receptors, Cannabinoid , Receptors, Drug/physiology
SELECTION OF CITATIONS
SEARCH DETAIL