Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Eur J Neurosci ; 51(3): 742-754, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31544297

RESUMO

Persons suffering from opioid use disorder (OUD) experience long-lasting dysphoric symptoms well into extended periods of withdrawal. This protracted withdrawal syndrome is notably characterized by heightened anxiety and hyperkatifeia. Here, we investigated if an exacerbated withdrawal model of acute morphine dependence results in lasting behavioral adaptation 6 weeks into forced abstinence in C57BL/6J mice. We found that our exacerbated morphine withdrawal paradigm produced distinct alterations in behavior in elevated plus maze (EPM), open field, and social interaction tests in male and female mice. Following protracted withdrawal male mice showed enhanced exploration of the open arms of the EPM, reduced latency to enter the corner of the OF, and a social interaction deficit. In contrast, female mice showed enhanced thigmotaxis in the OF. In both sexes, protracted withdrawal enhanced locomotor behavior in response to subsequent morphine challenge, albeit at different doses. These findings will be relevant for future investigation examining the neural mechanisms underlying these behaviors and will aid in uncovering physiological sex differences in response to opioid withdrawal.


Assuntos
Analgésicos Opioides , Síndrome de Abstinência a Substâncias , Analgésicos Opioides/uso terapêutico , Animais , Ansiedade , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina , Síndrome de Abstinência a Substâncias/tratamento farmacológico
2.
Addict Biol ; 25(3): e12748, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-30963693

RESUMO

The United States is experiencing an opioid crisis imposing enormous fiscal and societal costs and driving the staggering overdose death rate. While prescription opioid analgesics are essential for treating acute pain, cessation of use in individuals with a physical dependence induces an aversive withdrawal syndrome that promotes continued drug use to alleviate/avoid these symptoms. Additionally, repeated bouts of withdrawal often lead to an increased propensity for relapse. Understanding the neurobiology underlying withdrawal is essential for providing novel treatment options to alleviate physiological and affective components accompanying the cessation of opiate use. Here, we administered morphine and precipitated withdrawal with naloxone to investigate behavioral and cellular responses in C57BL/6J male and female mice. Following 3 days of administration, both male and female mice demonstrated sensitized withdrawal symptoms. Since the bed nucleus of the stria terminalis (BNST) plays a role in mediating withdrawal-associated behaviors, we examined plastic changes in inhibitory synaptic transmission within this structure 24 hours following the final precipitated withdrawal. In male mice, morphine withdrawal increased spontaneous GABAergic signaling compared with controls. In contrast, morphine withdrawal decreased spontaneous GABAergic signaling in female mice. Intriguingly, these opposing GABAergic effects were contingent upon activity-dependent dynamics within the ex vivo slice. Our findings suggest that male and female mice exhibit some divergent cellular responses in the BNST following morphine withdrawal, and alterations in BNST inhibitory signaling may contribute to the expression of behaviors following opioid withdrawal.


Assuntos
Analgésicos Opioides/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Inibição Neural/efeitos dos fármacos , Núcleos Septais/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos Endogâmicos C57BL , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Dependência de Morfina , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Patch-Clamp , Núcleos Septais/citologia , Núcleos Septais/metabolismo , Núcleos Septais/fisiopatologia , Síndrome de Abstinência a Substâncias/etiologia , Ácido gama-Aminobutírico/metabolismo
3.
J Neurosci ; 33(33): 13367-74, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23946394

RESUMO

Two of the most commonly used procedures to study the abuse-related effects of drugs in laboratory animals are intravenous drug self-administration and reinstatement of extinguished behavior previously maintained by drug delivery. Intravenous self-administration is widely accepted to model ongoing drug-taking behavior, whereas reinstatement procedures are accepted to model relapse to drug taking following abstinence. Previous studies indicate that 5-HT2A receptor antagonists attenuate the reinstatement of cocaine-maintained behavior but not cocaine self-administration in rodents. Although the abuse-related effects of cocaine have been closely linked to brain dopamine systems, no previous study has determined whether this dissociation is related to differential regulation of dopamine neurotransmission. To elucidate the neuropharmacological and neuroanatomical mechanisms underlying this phenomenon, we evaluated the effects of the selective 5-HT2A receptor antagonist M100907 on intravenous cocaine self-administration and drug- and cue-primed reinstatement in rhesus macaques (Macaca mulatta). In separate subjects, we evaluated the role of 5-HT2A receptors in cocaine-induced dopamine overflow in the nucleus accumbens (n = 4) and the caudate nucleus (n = 5) using in vivo microdialysis. Consistent with previous studies, M100907 (0.3 mg/kg, i.m.) significantly attenuated drug- and cue-induced reinstatement but had no significant effects on cocaine self-administration across a range of maintenance doses. Importantly, M100907 (0.3 mg/kg, i.m.) attenuated cocaine-induced (1.0 mg/kg, i.v.) dopamine overflow in the caudate nucleus but not in the nucleus accumbens. These data suggest that important abuse-related effects of cocaine are mediated by distinct striatal dopamine projection pathways.


Assuntos
Encéfalo/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Dopamina/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Feminino , Fluorbenzenos/farmacologia , Macaca mulatta , Microdiálise , Piperidinas/farmacologia , Antagonistas da Serotonina/farmacologia
4.
Mol Pharmacol ; 85(4): 640-50, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24499709

RESUMO

Psychostimulants, such as cocaine and amphetamines, act primarily through the monoamine neurotransmitters dopamine (DA), norepinephrine, and serotonin. Although stimulant addiction research has largely focused on DA, medication development efforts targeting the dopaminergic system have thus far been unsuccessful, leading to alternative strategies aimed at abating stimulant abuse. Noradrenergic compounds have shown promise in altering the behavioral effects of stimulants in rodents, nonhuman primates, and humans. In this review, we discuss the contribution of each adrenergic receptor (AR) subtype (α1, α2, and ß) to five stimulant-induced behaviors relevant to addiction: locomotor activity, conditioned place preference, anxiety, discrimination, and self-administration. AR manipulation has diverse effects on these behaviors; each subtype profoundly influences outcomes in some paradigms but is inconsequential in others. The functional neuroanatomy and intracellular signaling mechanisms underlying the impact of AR activation/blockade on these behaviors remain largely unknown, presenting a new frontier for research on psychostimulant-AR interactions.


Assuntos
Comportamento Aditivo/psicologia , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Receptores Adrenérgicos/fisiologia , Anfetamina/farmacologia , Animais , Ansiedade/diagnóstico , Ansiedade/psicologia , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Aditivo/etiologia , Cocaína/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/fisiologia , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos beta/fisiologia , Autoadministração , Transdução de Sinais
5.
J Pharmacol Exp Ther ; 350(1): 144-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24817036

RESUMO

Disulfiram has shown promise as a pharmacotherapy for cocaine dependence in clinical settings, although it has many targets, and the behavioral and molecular mechanisms underlying its efficacy are unclear. One of many biochemical actions of disulfiram is inhibition of dopamine ß-hydroxylase (DBH), the enzyme that converts dopamine (DA) to norepinephrine (NE) in noradrenergic neurons. Thus, disulfiram simultaneously reduces NE and elevates DA tissue levels in the brain. In rats, both disulfiram and the selective DBH inhibitor nepicastat block cocaine-primed reinstatement, a paradigm which is thought to model some aspects of drug relapse. This is consistent with some clinical results and supports the use of DBH inhibitors for the treatment of cocaine dependence. The present study was conducted to confirm and extend these results in nonhuman primates. Squirrel monkeys trained to self-administer cocaine were pretreated with disulfiram or nepicastat prior to cocaine-induced reinstatement sessions. Neither DBH inhibitor altered cocaine-induced reinstatement. Unexpectedly, nepicastat administered alone induced a modest reinstatement effect in squirrel monkeys, but not in rats. To investigate the neurochemical mechanisms underlying the behavioral results, the effects of DBH inhibition on extracellular DA were analyzed in the nucleus accumbens (NAc) using in vivo microdialysis in squirrel monkeys. Both DBH inhibitors attenuated cocaine-induced DA overflow in the NAc. Hence, the attenuation of cocaine-induced changes in accumbal DA neurochemistry was not associated with altered cocaine-seeking behavior. Overall, the reported behavioral effects of DBH inhibition in rodent models of relapse did not extend to nonhuman primates under the conditions used in the current studies.


Assuntos
Cocaína/administração & dosagem , Dopamina beta-Hidroxilase/deficiência , Dopamina/metabolismo , Imidazóis/farmacologia , Norepinefrina/deficiência , Núcleo Accumbens/metabolismo , Tionas/farmacologia , Animais , Doenças do Sistema Nervoso Autônomo , Comportamento Aditivo/induzido quimicamente , Cocaína/antagonistas & inibidores , Condicionamento Operante/efeitos dos fármacos , Dissulfiram/farmacologia , Dopamina beta-Hidroxilase/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Extinção Psicológica/efeitos dos fármacos , Masculino , Norepinefrina/antagonistas & inibidores , Núcleo Accumbens/efeitos dos fármacos , Ratos , Saimiri , Autoadministração
6.
J Pharmacol Exp Ther ; 342(1): 177-87, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22514333

RESUMO

Inhibitors of fatty acid amide hydrolase (FAAH) and anandamide (AEA) uptake, which limit the degradation of endogenous cannabinoids, have received interest as potential therapeutics for pain. There is also evidence that endogenous cannabinoids mediate the antinociceptive effects of opioids. Assays of pain-elicited and pain-suppressed behavior have been used to differentiate the effects of drugs that specifically alter nociception from drugs that alter nociception caused by nonspecific effects such as catalepsy or a general suppression of activity. Using such procedures, this study examines the effects of the direct cannabinoid type 1 (CB1) agonist (-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl)cyclohexanol (CP55940), the FAAH inhibitor cyclohexylcarbamic acid 3'-carbamoylbiphenyl-3-yl ester (URB597), and the AEA uptake inhibitor N-(4-hydroxyphenyl) arachidonylamide (AM404). Additional experiments examined these compounds in combination with morphine. CP55940 produced antinociception in assays of pain-elicited, but not pain-suppressed, behavior and disrupted responding in an assay of schedule-controlled behavior. URB597 and AM404 produced antinociception in assays of pain-elicited and pain-suppressed behavior in which acetic acid was the noxious stimulus, but had no effect on the hotplate and schedule-controlled responding. CP55940 in combination with morphine resulted in effects greater than those of morphine alone in assays of pain-elicited and scheduled-controlled behavior but not pain-suppressed behavior. URB597 in combination with morphine resulted in enhanced morphine effects in assays of pain-elicited and pain-suppressed behavior in which diluted acetic acid was the noxious stimulus, but did not alter morphine's effects on the hotplate or schedule-controlled responding. These studies suggest that, compared with direct CB1 agonists, manipulations of endogenous cannabinoid signaling have enhanced clinical potential; however, their effects depend on the type of noxious stimulus.


Assuntos
Moduladores de Receptores de Canabinoides/metabolismo , Morfina/farmacologia , Dor/tratamento farmacológico , Dor/metabolismo , Amidoidrolases/antagonistas & inibidores , Analgésicos/farmacologia , Animais , Ácidos Araquidônicos/antagonistas & inibidores , Ácidos Araquidônicos/farmacologia , Benzamidas/farmacologia , Carbamatos/farmacologia , Cicloexanóis/farmacologia , Endocanabinoides/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nociceptividade/efeitos dos fármacos , Alcamidas Poli-Insaturadas/antagonistas & inibidores , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo
7.
Pharmacol Biochem Behav ; 218: 173431, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35850178

RESUMO

Ovarian hormones influence the activity of endogenous opioids, and exogenous administration of estradiol reduces opioid intake and opioid seeking in animal models of opioid reward and reinforcement. The purpose of this study was to examine the effects of ovarian hormones on the discriminative stimulus effects of morphine and naloxone-precipitated opioid withdrawal. To this end, separate groups of ovariectomized female rats were trained to discriminate the stimulus effects of either 3.0 or 10 mg/kg morphine, and substitution tests were conducted with estradiol or progesterone alone and in combination with morphine. At the conclusion of discrimination testing, rats were treated chronically with estradiol, progesterone, or their combination, and challenged with naloxone to measure opioid-like withdrawal symptoms. Finally, the effects of estradiol, progesterone, and their combination were examined on naloxone-precipitated withdrawal in morphine-dependent rats. Neither estradiol nor progesterone substituted for the morphine discriminative stimulus, but estradiol significantly increased the potency of morphine in rats trained to discriminate 10 mg/kg but not 3 mg/kg morphine. When administered chronically, neither hormone nor their combination produced an opioid-like withdrawal syndrome following a naloxone challenge. Acute administration of estradiol, but not progesterone or a combination of estradiol and progesterone, significantly reduced naloxone-precipitated weight loss in morphine-dependent rats. These data indicate that estradiol influences the behavioral effects of morphine, possibly by increasing endogenous tone at mu opioid receptors.


Assuntos
Dependência de Morfina , Síndrome de Abstinência a Substâncias , Transtornos Relacionados ao Uso de Substâncias , Analgésicos Opioides/farmacologia , Animais , Estradiol/farmacologia , Feminino , Morfina/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Progesterona/farmacologia , Ratos , Receptores Opioides mu
8.
Exp Clin Psychopharmacol ; 30(2): 127-131, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33001695

RESUMO

Heroin intake decreases markedly during proestrus in normally cycling female rats; however, it is not known whether estradiol, progesterone, or both hormones are responsible for these decreases in heroin intake. The purpose of the present study was to examine the roles of estradiol and progesterone in heroin intake by artificially inducing a proestrus state in ovariectomized rats. To this end, ovariectomized female rats were implanted with intravenous catheters and trained to self-administer heroin (0.0075 mg/kg/infusion) on a fixed ratio (FR1) schedule of reinforcement. After 1 week of training, rats were tested at weekly intervals with estradiol (0.005 mg, sc) or vehicle 22 hr before a test session and progesterone (0.125 mg, sc) or vehicle 0.5 hr before a test session to artificially mimic the naturally occurring hormone concentrations characteristic of late proestrus. Administration of estradiol 22 hr prior to testing and progesterone 0.5 hr prior to testing significantly reduced heroin intake relative to the previous training day and vehicle control. It is interesting that this same effect was observed when only estradiol, but not progesterone, was administered. These data suggest that estradiol but not progesterone is responsible for the proestrus-induced decreases in heroin intake previously reported in normally cycling female rats. These findings differ from those reported previously with stimulants and suggest that estrogen-based pharmacotherapies may be of value to women with opioid use disorder. (PsycInfo Database Record (c) 2022 APA, all rights reserved).


Assuntos
Estradiol , Heroína , Animais , Estradiol/farmacologia , Feminino , Heroína/farmacologia , Humanos , Proestro , Progesterona/farmacologia , Ratos
9.
Behav Pharmacol ; 22(5-6): 540-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21712708

RESUMO

This study explored the involvement of N-methyl-D-aspartate (NMDA) in the effects of µ-opioid agonists. A hot-plate procedure was used to assess antinociception and tolerance in mice in which the NR1 subunit of the NMDA receptor was reduced [knockdown (KD)] to approximately 10%, and in mice treated with the NMDA antagonist, (-)-6-phosphonomethyl-deca-hydroisoquinoline-3-carboxylic acid (LY235959). The µ opioid agonists, morphine, l-methadone and fentanyl, were approximately three-fold less potent in the NR1 KD mice than in wild-type (WT) controls; however, the development of morphine tolerance and dependence did not differ markedly in the NR1 KD and the WT mice. Acute administration of the NMDA antagonist, LY235959, produced dose-dependent, leftward shifts in the morphine dose-effect curve in the WT mice, but not in the NR1 KD mice. Chronic administration of LY235959 during the morphine tolerance regimen did not attenuate the development of tolerance in the NR1 KD or the WT mice. These results indicate that the NR1 subunit of the NMDA receptor does not play a prominent role in µ opioid tolerance.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Dor/tratamento farmacológico , Receptores de N-Metil-D-Aspartato/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/farmacologia , Técnicas de Silenciamento de Genes , Isoquinolinas/administração & dosagem , Isoquinolinas/farmacologia , Masculino , Camundongos , Morfina/administração & dosagem , Dependência de Morfina/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Receptores Opioides mu/agonistas
10.
Behav Brain Res ; 409: 113329, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-33933523

RESUMO

Heroin intake decreases during the proestrus phase of the estrous cycle in female, Long-Evans rats. The purpose of this study was to (1) determine if proestrus-associated decreases in heroin intake extend across rat strains and (2) determine if proestrus-associated decreases in responding extend to a nondrug reinforcer. Female rats were implanted with intravenous catheters and trained to self-administer heroin. Estrous cycle was tracked daily for the duration of the study. During testing, Lewis, Sprague Dawley, and Long-Evans rats self-administered low (0.0025 mg/kg) and high (0.0075 mg /kg) doses of heroin and then self-administered sugar on fixed ratio (FR1) schedules of reinforcement. Heroin intake decreased significantly during proestrus in all three rat strains under at least one dose condition; however, sugar intake did not decrease during proestrus in any strain. These data suggest that responding maintained by heroin, but not a nondrug reinforcer, significantly decreases during proestrus in female rats and that these effects are consistent across rat strain.


Assuntos
Comportamento Animal/fisiologia , Açúcares da Dieta/administração & dosagem , Ciclo Estral/fisiologia , Heroína/administração & dosagem , Entorpecentes/administração & dosagem , Animais , Ciclo Estral/metabolismo , Feminino , Ratos , Ratos Endogâmicos Lew , Ratos Long-Evans , Ratos Sprague-Dawley , Esquema de Reforço , Autoadministração , Açúcares
11.
Drug Alcohol Depend ; 225: 108816, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34171824

RESUMO

Heroin intake decreases significantly during proestrus in normally cycling female rats, and this effect is mediated by endogenous estradiol but not endogenous progesterone. The purpose of this study was to determine whether chronic administration of exogenous estradiol decreases intake of the semi-synthetic opioid, heroin, and the fully synthetic opioid, remifentanil, in intact female rats. Normally cycling female rats were implanted with intravenous catheters and trained to self-administer heroin on a fixed ratio (FR1) schedule of reinforcement. Rats were treated chronically with daily administration of either a low dose of estradiol (0.5 mcg, sc), a high dose of estradiol (5.0 mcg, sc), or vehicle (peanut oil, sc). After two weeks of heroin self-administration training, dose-effect curves were determined for both heroin and remifentanil. Chronic administration of estradiol non-significantly decreased heroin intake and significantly decreased remifentanil intake. Estradiol-induced decreases in remifentanil intake were dose-dependent, characterized by large effect sizes, and greatest in rats treated with the high dose of estradiol. These data indicate that chronic estradiol administration decreases opioid intake in intact female rats with medium to large effect sizes across opioids. These findings suggest that estrogen-based pharmacotherapies may represent a novel treatment approach for women with opioid use disorder.


Assuntos
Analgésicos Opioides , Estradiol , Animais , Relação Dose-Resposta a Droga , Estrogênios/farmacologia , Feminino , Heroína , Ratos , Ratos Sprague-Dawley , Autoadministração
12.
Eur J Pharmacol ; 913: 174646, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34800468

RESUMO

3,4-Methylenedioxymethamphetamine (MDMA) is classified as an entactogen, producing feelings of emotional openness and relatedness. One unique feature of MDMA is that people tend to selectively take this drug in social and/or intimate situations. Although MDMA is recognized as having abuse liability, preclinical studies report that it has weak reinforcing effects in animals. The objective of this study was to characterize the positive reinforcing and prosocial effects of MDMA in a translational model of the social environment in which two rats have simultaneous and contingent access to MDMA in close physical proximity. To this end, MDMA self-administration was examined on both fixed and progressive ratio schedules of reinforcement in six groups of rats: (1) isolated males, (2) isolated females, (3) male-male dyads, (4) female-female dyads, (5) male-female dyads, and (6) female-male dyads. For pair-housed rats, data from both rats were analyzed. Next, social preferences were examined in a partner preference test. MDMA failed to produce positive reinforcing effects under all conditions examined. Across a 30-fold dose range (0.01-1.0 mg/kg/infusion), MDMA did not maintain higher responding than saline on both schedules of reinforcement and in all groups tested. In partner preference tests, a history of shared exposure to MDMA did not establish a social preference, and acute administration of MDMA failed to establish a preference for another MDMA-treated rat. These data suggest that social contact does not increase the positive reinforcing effects of MDMA in rats, and that neither contingent nor noncontingent MDMA administration establishes a social preference in rats.


Assuntos
N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Reforço Psicológico , Interação Social/efeitos dos fármacos , Animais , Feminino , Masculino , Modelos Animais , Ratos , Autoadministração
13.
Psychopharmacology (Berl) ; 238(4): 969-978, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404736

RESUMO

RATIONALE: Heroin intake decreases during the proestrus phase of the estrous cycle in female rats. Circulating concentrations of both estradiol and progesterone peak during proestrus, and it is not known which of these hormones, or their combination, are responsible for these effects. OBJECTIVES: The purpose of this study was to determine the effects of estradiol, progesterone, and their combination on heroin self-administration in female rats. METHODS: In Experiment 1, the estrous cycle of intact female rats was tracked daily. If a rat was in proestrus, either the estrogen receptor antagonist, raloxifene, the progesterone receptor antagonist, mifepristone, or their combination was administered 30 min prior to a heroin self-administration session. In Experiment 2, separate groups of ovariectomized female rats were treated chronically with exogenous estradiol, progesterone, estradiol + progesterone, or vehicle, and heroin intake was examined over a 100-fold dose range. RESULTS: In Experiment 1, raloxifene, but not mifepristone, significantly blocked proestrus-associated decreases in heroin intake. In Experiment 2, estrogentreated rats self-administered less heroin than any other group and significantly less heroin than rats treated with progesterone. CONCLUSIONS: These data suggest that (1) estradiol but not progesterone is responsible for proestrus-associated decreases in heroin intake and (2) estradiol decreases heroin intake relative to progesterone. These data differ from those reported previously with stimulants and suggest that estrogen-based pharmacotherapies may be of value to women with opioid use disorder.


Assuntos
Estradiol/administração & dosagem , Heroína/administração & dosagem , Progesterona/administração & dosagem , Animais , Estradiol/farmacologia , Estro/fisiologia , Feminino , Heroína/farmacologia , Mifepristona/administração & dosagem , Mifepristona/farmacologia , Proestro/fisiologia , Progesterona/farmacologia , Ratos , Ratos Long-Evans
14.
Neuron ; 109(8): 1365-1380.e5, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33740416

RESUMO

Sex differences in pain severity, response, and pathological susceptibility are widely reported, but the neural mechanisms that contribute to these outcomes remain poorly understood. Here we show that dopamine (DA) neurons in the ventrolateral periaqueductal gray/dorsal raphe (vlPAG/DR) differentially regulate pain-related behaviors in male and female mice through projections to the bed nucleus of the stria terminalis (BNST). We find that activation of vlPAG/DRDA+ neurons or vlPAG/DRDA+ terminals in the BNST reduces nociceptive sensitivity during naive and inflammatory pain states in male mice, whereas activation of this pathway in female mice leads to increased locomotion in the presence of salient stimuli. We additionally use slice physiology and genetic editing approaches to demonstrate that vlPAG/DRDA+ projections to the BNST drive sex-specific responses to pain through DA signaling, providing evidence of a novel ascending circuit for pain relief in males and contextual locomotor response in females.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Núcleo Dorsal da Rafe/fisiologia , Atividade Motora/fisiologia , Dor/fisiopatologia , Substância Cinzenta Periaquedutal/fisiologia , Caracteres Sexuais , Animais , Comportamento Animal/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Medição da Dor
15.
Behav Pharmacol ; 20(4): 312-21, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19584714

RESUMO

Environmental enrichment produces functional changes in mesolimbic dopamine transmission and alters sensitivity to psychomotor stimulants. These manipulations also alter the control rate of many behaviors that are sensitive to stimulant administration, which can make comparison of drug effects between isolated and enriched subjects difficult. The purpose of this study was to examine the effects of environmental enrichment on control rates of behavior and on sensitivity to cocaine in tests of locomotor activity, drug self-administration, conditioned place preference, and toxicity. In the locomotor activity test, isolated rats exhibited greater activity after the administration of cocaine, but also had higher control rates of activity. When locomotor activity was expressed as a percentage of saline control values, enriched rats exhibited a greater increase relative to their own control than isolated rats. In the drug self-administration procedure, isolated rats had higher breakpoints on a progressive-ratio schedule of reinforcement when responding was maintained by cocaine; however, isolated rats also had higher breakpoints in saline substitution tests and higher rates of inactive lever responding. When the self-administration data were expressed as a percentage of these control values, enriched rats exhibited a greater increase in responding relative to their own control rates than isolated rats. No differences were observed between isolated and enriched rats under control conditions in the place preference and toxicity studies. In both of these procedures, enriched rats were more sensitive than isolated rats to all the doses of cocaine tested. These data emphasize the importance of considering control rates of behavior in studies examining environmental enrichment and drug sensitivity, and suggest that environmental enrichment increases sensitivity to cocaine across a range of dependent measures when differences in control rates of behavior are taken into account.


Assuntos
Comportamento Animal/efeitos dos fármacos , Cocaína/farmacologia , Atividade Motora/efeitos dos fármacos , Entorpecentes/farmacologia , Meio Social , Isolamento Social , Animais , Cocaína/administração & dosagem , Feminino , Entorpecentes/administração & dosagem , Ratos , Ratos Long-Evans , Autoadministração
16.
ACS Chem Neurosci ; 10(4): 1872-1874, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30920796

RESUMO

To overcome the limitations of fast scan cyclic voltammetry (FSCV) to discriminate between catecholamines, we discuss new approaches to monitor the dynamics of these neurochemicals with high spatial, genetic, and temporal specificity.


Assuntos
Catecolaminas/análise , Dopamina/análise , Técnicas Eletroquímicas/tendências , Aprendizado de Máquina/tendências , Norepinefrina/análise , Optogenética/tendências , Animais , Catecolaminas/metabolismo , Dopamina/metabolismo , Técnicas Eletroquímicas/métodos , Humanos , Norepinefrina/metabolismo , Optogenética/métodos
17.
Neuropsychopharmacology ; 44(10): 1728-1741, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31141817

RESUMO

Kappa opioid receptor (KOR) agonists show promise in ameliorating disorders, such as addiction and chronic pain, but are limited by dysphoric and aversive side effects. Clinically beneficial effects of KOR agonists (e.g., analgesia) are predominantly mediated by heterotrimeric G protein signaling, whereas ß-arrestin signaling is considered central to their detrimental side effects (e.g., dysphoria/aversion). Here we show that Regulator of G protein Signaling-12 (RGS12), via independent signaling mechanisms, simultaneously attenuates G protein signaling and augments ß-arrestin signaling downstream of KOR, exhibiting considerable selectivity in its actions for KOR over other opioid receptors. We previously reported that RGS12-null mice exhibit increased dopamine transporter-mediated dopamine (DA) uptake in the ventral (vSTR), but not dorsal striatum (dSTR), as well as reduced psychostimulant-induced hyperlocomotion; in the current study, we found that these phenotypes are reversed following KOR antagonism. Fast-scan cyclic voltammetry studies of dopamine (DA) release and reuptake suggest that striatal disruptions to KOR-dependent DAergic neurotransmission in RGS12-null mice are restricted to the nucleus accumbens. In both ventral striatal tissue and transfected cells, RGS12 and KOR are seen to interact within a protein complex. Ventral striatal-specific increases in KOR levels and KOR-induced G protein activation are seen in RGS12-null mice, as well as enhanced sensitivity to KOR agonist-induced hypolocomotion and analgesia-G protein signaling-dependent behaviors; a ventral striatal-specific increase in KOR levels was also observed in ß-arrestin-2-deficient mice, highlighting the importance of ß-arrestin signaling to establishing steady-state KOR levels in this particular brain region. Conversely, RGS12-null mice exhibited attenuated KOR-induced conditioned place aversion (considered a ß-arrestin signaling-dependent behavior), consistent with the augmented KOR-mediated ß-arrestin signaling seen upon RGS12 over-expression. Collectively, our findings highlight a role for RGS12 as a novel, differential regulator of both G protein-dependent and -independent signaling downstream of KOR activation.


Assuntos
Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Proteínas RGS/genética , Receptores Opioides kappa/metabolismo , Estriado Ventral/metabolismo , beta-Arrestinas/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Leucina Encefalina-2-Alanina/farmacologia , Feminino , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Núcleo Accumbens/efeitos dos fármacos , Receptores Opioides kappa/agonistas , Transdução de Sinais , Transmissão Sináptica/efeitos dos fármacos , Estriado Ventral/efeitos dos fármacos
18.
ACS Chem Neurosci ; 10(4): 1908-1914, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30252438

RESUMO

Stress can drive adaptive changes to maintain survival during threatening stimuli. Chronic stress exposure, however, may result in pathological adaptations. A key neurotransmitter involved in stress signaling is norepinephrine. Previous studies show that acute stress elevates norepinephrine levels in the bed nucleus of the stria terminalis (BNST), a critical node regulating anxiety and upstream of stress responses. Here, we use mice expressing channelrhodopsin in norepinephrine neurons to selectively activate terminals in the BNST, and measure norepinephrine release with optogenetics-assisted fast-scan cyclic voltammetry (FSCV). We demonstrate that while corticosterone habituates to chronic restraint stress, cFos activation of medullary norepinephrine neurons shows equivalent activation under both acute and chronic stress conditions. Mice exposed to a single restraint session show an identical optically stimulated norepinephrine release profile compared to that of unexposed mice. Mice experiencing 5 days of restraint stress, however, show elevated norepinephrine release across multiple stimulation parameters, and reduced sensitivity to the α2-adrenergic receptor (AR) antagonist idazoxan. These data are the first to examine norepinephrine release in the BNST to tonic and phasic stimulation frequencies, and confirm that repeated stress alters autoreceptor sensitivity.


Assuntos
Norepinefrina/metabolismo , Optogenética/métodos , Núcleos Septais/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Estimulação Luminosa/métodos , Núcleos Septais/química
19.
Drug Alcohol Depend ; 98(1-2): 129-35, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18585870

RESUMO

Aerobic exercise can serve as an alternative, non-drug reinforcer in laboratory animals and has been recommended as a potential intervention for substance abusing populations. Unfortunately, relatively little empirical data have been collected that specifically address the possible protective effects of voluntary, long-term exercise on measures of drug self-administration. The purpose of the present study was to examine the effects of chronic exercise on sensitivity to the positive-reinforcing effects of cocaine in the drug self-administration procedure. Female rats were obtained at weaning and immediately divided into two groups. Sedentary rats were housed individually in standard laboratory cages that permitted no exercise beyond normal cage ambulation; exercising rats were housed individually in modified cages equipped with a running wheel. After 6 weeks under these conditions, rats were surgically implanted with venous catheters and trained to self-administer cocaine on a fixed-ratio schedule of reinforcement. Once self-administration was acquired, cocaine was made available on a progressive ratio schedule and breakpoints were obtained for various doses of cocaine. Sedentary and exercising rats did not differ in the time to acquire cocaine self-administration or responding on the fixed-ratio schedule of reinforcement. However, on the progressive ratio schedule, breakpoints were significantly lower in exercising rats than sedentary rats when responding was maintained by both low (0.3mg/kg/infusion) and high (1.0mg/kg/infusion) doses of cocaine. In exercising rats, greater exercise output prior to catheter implantation was associated with lower breakpoints at the high dose of cocaine. These data indicate that chronic exercise decreases the positive-reinforcing effects of cocaine and support the possibility that exercise may be an effective intervention in drug abuse prevention and treatment programs.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/psicologia , Cocaína/administração & dosagem , Motivação , Condicionamento Físico Animal/psicologia , Reforço Psicológico , Animais , Peso Corporal , Relação Dose-Resposta a Droga , Feminino , Infusões Intravenosas , Ratos , Ratos Long-Evans , Esquema de Reforço , Autoadministração
20.
Physiol Behav ; 94(4): 563-8, 2008 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-18456292

RESUMO

Several studies report that environmental enrichment enhances sensitivity to opioid receptor agonists in male rats. Very few studies have examined the effects of enrichment in female rats, and thus it is not clear whether females are similarly sensitive to these effects. Consequently, the purpose of the present study was to examine the effects of environmental enrichment on sensitivity to representative mu, kappa, and mixed-action opioids in female rats. Following a protocol established in males, females were obtained at weaning and randomly assigned to two groups immediately upon arrival: isolated rats were housed individually with no visual or tactile contact with other rats; enriched rats were housed in groups of four in large cages and given various novel objects on a regular basis. After 6 weeks under these conditions, the antinociceptive effects of mu (morphine, levorphanol), kappa (spiradoline, U69,593), and mixed-action (buprenorphine, butorphanol) opioids were examined in a warm-water, tail-withdrawal procedure. All the opioids examined produced dose-dependent increases in antinociception; however, no differences in opioid sensitivity were observed between the two groups. To determine whether these findings were consistent across behavioral endpoints, the antidiuretic effects of representative mu opioids, and the diuretic effects of representative kappa opioids, were examined in female rats reared under isolated or enriched conditions for 10 weeks. Similar to that seen in the antinociceptive experiment, no significant differences in opioid sensitivity were observed between groups. These data indicate that environmental enrichment does not alter sensitivity to the effects of opioid receptor agonists in female rats, and suggest that females may respond differently to environmental enrichment than males.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Animal/efeitos dos fármacos , Meio Ambiente , Comportamento Exploratório/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Analgésicos Opioides/classificação , Animais , Antidiuréticos/farmacologia , Condicionamento Operante/efeitos dos fármacos , Diuréticos/farmacologia , Feminino , Ratos , Ratos Long-Evans , Receptores Opioides kappa/fisiologia , Receptores Opioides mu/fisiologia , Isolamento Social
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA