Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Psychiatry ; 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38838841

RESUMO

BACKGROUND: Past research illuminated pivotal roles of dopamine D3 receptors (D3Rs) in the rewarding effects of cocaine and opioids. However, the cellular and neural circuit mechanisms underlying these actions remain unclear. METHODS: We employed Cre-LoxP techniques to selectively delete D3R from presynaptic dopamine neurons or postsynaptic dopamine D1R-expressing neurons in male and female mice. We utilized RNAscope in situ hybridization, immunohistochemistry, RT-PCR, voltammetry, optogenetics, microdialysis, and behavioral assays (n≥8) to functionally characterize the roles of presynaptic versus postsynaptic D3Rs in cocaine and opioid actions. RESULTS: Our results revealed D3R expression in ∼20% of midbrain dopamine neurons and ∼70% of D1R-expressing neurons in the nucleus accumbens. While D2R was expressed in ∼80% dopamine neurons, we found no D2R and D3R colocalization among these cells. Selective deletion of D3Rs from dopamine neurons increased exploratory behavior in novel environments and enhanced pulse-evoked NAc dopamine release. Conversely, D3R deletion from D1R-expressing neurons attenuated locomotor responses to D1-like and D2-like agonists. Strikingly, D3R deletion from either cell type reduced oxycodone self-administration and oxycodone-enhanced brain-stimulation reward. In contrast, neither of these D3R deletions impacted cocaine self-administration, cocaine-enhanced brain-stimulation reward, or cocaine-induced hyperlocomotion. Furthermore, D3R knockout in dopamine neurons reduced oxycodone-induced hyperactivity and analgesia, while deletion from D1R-expressing neurons potentiated opioid-induced hyperactivity without affecting analgesia. CONCLUSIONS: We dissected presynaptic versus postsynaptic D3R function in the mesolimbic dopamine system. D2R and D3R are expressed in different populations of midbrain dopamine neurons, regulating dopamine release. The mesolimbic D3Rs are critically involved in the actions of opioids but not cocaine.

2.
Mol Psychiatry ; 28(10): 4203-4214, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37479780

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors that regulate gene expression. Δ9-tetrahydrocannabinol (Δ9-THC) is a PPARγ agonist and some endocannabinoids are natural activators of PPARα and PPARγ. However, little is known regarding their cellular distributions in the brain and functional roles in cannabinoid action. Here, we first used RNAscope in situ hybridization and immunohistochemistry assays to examine the cellular distributions of PPARα and PPARγ expression in the mouse brain. We found that PPARα and PPARγ are expressed in ~70% of midbrain dopamine (DA) neurons. In the amygdala, PPARα is expressed in ~60% of glutamatergic neurons, while PPARγ is expressed in ~60%  of GABA neurons. However, no PPARα/γ signal was detected in GABA neurons in the nucleus accumbens. We then used a series of behavioral assays to determine the functional roles of PPARα/γ in the CNS effects of Δ9-THC. We found that optogenetic stimulation of midbrain DA neurons was rewarding as assessed by optical intracranial self-stimulation (oICSS) in DAT-cre mice. Δ9-THC and a PPARγ (but not PPARα) agonist dose-dependently inhibited oICSS. Pretreatment with PPARα or PPARγ antagonists attenuated the Δ9-THC-induced reduction in oICSS and Δ9-THC-induced anxiogenic effects. In addition, a PPARγ agonist increased, while PPARα or PPARγ antagonists decreased open-field locomotion. Pretreatment with PPARα or PPARγ antagonists potentiated Δ9-THC-induced hypoactivity and catalepsy but failed to alter Δ9-THC-induced analgesia, hypothermia and immobility. These findings provide the first anatomical and functional evidence supporting an important role of PPARα/γ in DA-dependent behavior and cannabinoid action.


Assuntos
Canabinoides , PPAR alfa , Camundongos , Animais , PPAR alfa/metabolismo , Dopamina , Canabinoides/farmacologia , PPAR gama/metabolismo , Dronabinol , Neurônios Dopaminérgicos/metabolismo , Mesencéfalo/metabolismo
3.
Res Sq ; 2023 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-36909477

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors that regulate gene expression. Δ 9 -tetrahydrocannabinol (Δ 9 -THC) is a PPARg agonist and some endocannabinoids are natural activators of PPAR a and PPARg. Therefore, both the receptors are putative cannabinoid receptors. However, little is known regarding their cellular distributions in the brain and functional roles in cannabinoid action. Here we first used RNAscope in situ hybridization and immunohistochemistry assays to examine the cellular distributions of PPARα and PPARγ expression in the mouse brain. We found that PPARα and PPARγ are highly expressed in ~70% midbrain dopamine (DA) neurons and in ~50% GABAergic and ~50% glutamatergic neurons in the amygdala. However, no PPARα/γ signal was detected in GABAergic neurons in the nucleus accumbens. We then used a series of behavioral assays to determine the functional roles of PPARα/γ in the CNS effects of Δ 9 -THC. We found that optogenetic stimulation of midbrain DA neurons was rewarding as assessed by optical intracranial self-stimulation (oICSS) in DAT-cre mice. Δ 9 -THC and a PPARγ (but not PPARα) agonist dose-dependently inhibited oICSS, suggesting that dopaminergic PPARγ modulates DA-dependent behavior. Surprisingly, pretreatment with PPARα or PPARγ antagonists dose-dependently attenuated the Δ 9 -THC-induced reduction in oICSS and anxiogenic effects. In addition, a PPARγ agonist increased, while PPARa or PPARγ antagonists decreased open-field locomotion. Pretreatment with PPARa or PPARγ antagonists potentiated Δ 9 -THC-induced hypoactivity and catalepsy but failed to alter Δ 9 -THC-induced analgesia, hypothermia and immobility. These findings provide the first anatomical and functional evidence supporting an important role of PPARa/g in DA-dependent behavior and cannabinoid action.

4.
J Neurosci ; 43(3): 373-385, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36517243

RESUMO

Cannabinoids modulate dopamine (DA) transmission and DA-related behavior, which has been thought to be mediated initially by activation of cannabinoid CB1 receptors (CB1Rs) on GABA neurons. However, there is no behavioral evidence supporting it. In contrast, here we report that CB1Rs are also expressed in a subset of DA neurons and functionally underlie cannabinoid action in male and female mice. RNAscope in situ hybridization (ISH) assays demonstrated CB1 mRNA in tyrosine hydroxylase (TH)-positive DA neurons in the ventral tegmental area (VTA) and glutamate decarboxylase 1 (GAD1)-positive GABA neurons. The CB1R-expressing DA neurons were located mainly in the middle portion of the VTA with the number of CB1-TH colocalization progressively decreasing from the medial to the lateral VTA. Triple-staining assays indicated CB1R mRNA colocalization with both TH and vesicular glutamate transporter 2 (VgluT2, a glutamate neuronal marker) in the medial VTA close to the midline of the brain. Optogenetic activation of this population of DA neurons was rewarding as assessed by optical intracranial self-stimulation. Δ9-tetrahydrocannabinol (Δ9-THC) or ACEA (a selective CB1R agonist) dose-dependently inhibited optical intracranial self-stimulation in DAT-Cre control mice, but not in conditional knockout mice with the CB1R gene absent in DA neurons. In addition, deletion of CB1Rs from DA neurons attenuated Δ9-THC-induced reduction in DA release in the NAc, locomotion, and anxiety. Together, these findings indicate that CB1Rs are expressed in a subset of DA neurons that corelease DA and glutamate, and functionally underlie cannabinoid modulation of DA release and DA-related behavior.SIGNIFICANCE STATEMENT Cannabinoids produce a series of psychoactive effects, such as aversion, anxiety, and locomotor inhibition in rodents. However, the cellular and receptor mechanisms underlying these actions are not fully understood. Here we report that CB1 receptors are expressed not only in GABA neurons but also in a subset of dopamine neurons, which are located mainly in the medial VTA close to the midline of the midbrain and corelease dopamine and glutamate. Optogenetic activation of these dopamine neurons is rewarding, which is dose-dependently inhibited by cannabinoids. Selective deletion of CB1 receptor from dopamine neurons blocked cannabinoid-induced aversion, hypoactivity, and anxiolytic effects. These findings demonstrate that dopaminergic CB1 receptors play an important role in mediating cannabinoid action.


Assuntos
Ansiolíticos , Canabinoides , Feminino , Camundongos , Masculino , Animais , Canabinoides/farmacologia , Neurônios Dopaminérgicos/fisiologia , Ansiolíticos/farmacologia , Dronabinol/farmacologia , Dopamina/fisiologia , Receptores de Canabinoides , Área Tegmentar Ventral/fisiologia , Receptores Dopaminérgicos , Camundongos Knockout , Ácido Glutâmico/farmacologia , RNA Mensageiro , Receptor CB1 de Canabinoide/genética
5.
Sci Adv ; 8(35): eabo1440, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36054363

RESUMO

Physical exercise is rewarding and protective against drug abuse and addiction. However, the neural mechanisms underlying these actions remain unclear. Here, we report that long-term wheel-running produced a more robust increase in c-fos expression in the red nucleus (RN) than in other brain regions. Anatomic and functional assays demonstrated that most RN magnocellular portion (RNm) neurons are glutamatergic. Wheel-running activates a subset of RNm glutamate neurons that project to ventral tegmental area (VTA) dopamine neurons. Optogenetic stimulation of this pathway was rewarding, as assessed by intracranial self-stimulation and conditioned place preference, whereas optical inhibition blocked wheel-running behavior. Running wheel access decreased cocaine self-administration and cocaine seeking during extinction. Last, optogenetic stimulation of the RNm-to-VTA glutamate pathway inhibited responding to cocaine. Together, these findings indicate that physical exercise activates a specific RNm-to-VTA glutamatergic pathway, producing exercise reward and reducing cocaine intake.

6.
Neuropsychopharmacology ; 47(13): 2309-2318, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35879349

RESUMO

The non-medical use of opioids has become a national crisis in the USA. Developing non-opioid pharmacotherapies for controlling this opioid epidemic is urgent. Dopamine D3 receptor (D3R) antagonists and low efficacy partial agonists have shown promising profiles in animal models of opioid use disorders (OUD). However, to date, advancement to human studies has been limited. Here we report the effects of (S)- and (R)-enantiomers of (±)-ABS01-113, structural analogs of the D3R partial agonist, (±)-VK4-40, in which the 3-OH in the linking chain is replaced by 3-F group. (S)- and (R)-ABS01-113 are identical in chemical structure but with opposite chirality. In vitro receptor binding and functional assays indicate that (S)-ABS01-113 is an efficacious (55%) and potent (EC50 = 7.6 ± 3.9 nM) D3R partial agonist, while the (R)-enantiomer is a potent D3R antagonist (IC50 = 11.4 nM). Both (S)- and (R)-ABS01-113 bind with high affinity to D3R (Ki = 0.84 ± 0.16 and 0.37 ± 0.06 nM, respectively); however, the (S)-enantiomer is more D3/D2-selective (>1000-fold). Pharmacokinetic analyses indicate that both enantiomers display excellent oral bioavailability and high brain penetration. Systemic administration of (S)- or (R)-ABS01-113 alone failed to alter open-field locomotion in male rats and mice. Interestingly, pretreatment with (S)- or (R)-ABS01-113 attenuated heroin-enhanced hyperactivity, heroin self-administration, and (heroin + cue)-induced reinstatement of drug-seeking behavior. Together, these findings reveal that both enantiomers, particularly the highly selective and efficacious D3R partial agonist (S)-ABS01-113, demonstrate promising translational potential for the treatment of OUD.


Assuntos
Transtornos Relacionados ao Uso de Opioides , Receptores de Dopamina D3 , Animais , Ratos , Masculino , Camundongos , Humanos , Receptores de Dopamina D3/metabolismo , Heroína , Antagonistas de Dopamina/farmacologia , Comportamento de Procura de Droga , Analgésicos Opioides/farmacologia , Agonistas de Dopamina/farmacologia
7.
Transl Psychiatry ; 12(1): 286, 2022 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-35851573

RESUMO

Cannabinoid CB1 receptors (CB1Rs) have been major targets in medication development for the treatment of substance use disorders. However, clinical trials with rimonabant, a CB1R antagonist/inverse agonist, failed due to severe side effects. Here, we evaluated the therapeutic potential of PIMSR, a neutral CB1R antagonist lacking an inverse agonist profile, against cocaine's behavioral effects in experimental animals. We found that systemic administration of PIMSR dose-dependently inhibited cocaine self-administration under fixed-ratio (FR5), but not FR1, reinforcement, shifted the cocaine self-administration dose-response curve downward, decreased incentive motivation to seek cocaine under progressive-ratio reinforcement, and reduced cue-induced reinstatement of cocaine seeking. PIMSR also inhibited oral sucrose self-administration. Importantly, PIMSR alone is neither rewarding nor aversive as assessed by place conditioning. We then used intracranial self-stimulation (ICSS) to explore the possible involvement of the mesolimbic dopamine system in PIMSR's action. We found that PIMSR dose-dependently attenuated cocaine-enhanced ICSS maintained by electrical stimulation of the medial forebrain bundle in rats. PIMSR itself failed to alter electrical ICSS, but dose-dependently inhibited ICSS maintained by optical stimulation of midbrain dopamine neurons in transgenic DAT-Cre mice, suggesting the involvement of dopamine-dependent mechanisms. Lastly, we examined the CB1R mechanisms underlying PIMSR's action. We found that PIMSR pretreatment attenuated Δ9-tetrahydrocannabinol (Δ9-THC)- or ACEA (a selective CB1R agonist)-induced reduction in optical ICSS. Together, our findings suggest that the neutral CB1R antagonist PIMSR deserves further research as a promising pharmacotherapeutic for cocaine use disorder.


Assuntos
Cocaína , Transtornos Relacionados ao Uso de Substâncias , Animais , Comportamento Animal , Cocaína/farmacologia , Condicionamento Operante/fisiologia , Dopamina , Relação Dose-Resposta a Droga , Dronabinol/farmacologia , Camundongos , Ratos , Receptor CB1 de Canabinoide , Autoadministração
8.
Adv Pharmacol ; 93: 275-333, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35341569

RESUMO

Cannabis legalization continues to progress in many US states and other countries. Δ9-tetrahydrocannabinol (Δ9-THC) is the major psychoactive constituent in cannabis underlying both its abuse potential and the majority of therapeutic applications. However, the neural mechanisms underlying cannabis action are not fully understood. In this chapter, we first review recent progress in cannabinoid receptor research, and then examine the acute CNS effects of Δ9-THC or other cannabinoids (WIN55212-2) with a focus on their receptor mechanisms. In experimental animals, Δ9-THC or WIN55212-2 produces classical pharmacological effects (analgesia, catalepsy, hypothermia, hypolocomotion), biphasic changes in affect (reward vs. aversion, anxiety vs. anxiety relief), and cognitive deficits (spatial learning and memory, short-term memory). Accumulating evidence indicates that activation of CB1Rs underlies the majority of Δ9-THC or WIN55121-2's pharmacological and behavioral effects. Unexpectedly, glutamatergic CB1Rs preferentially underlie cannabis action relative to GABAergic CB1Rs. Functional roles for CB1Rs expressed on astrocytes and mitochondria have also been uncovered. In addition, Δ9-THC or WIN55212-2 is an agonist at CB2R, GPR55 and PPARγ receptors and recent studies implicate these receptors in a number of their CNS effects. Other receptors (such as serotonin, opioid, and adenosine receptors) also modulate Δ9-THC's actions and their contributions are detailed. This chapter describes the neural mechanisms underlying cannabis action, which may lead to new discoveries in cannabis-based medication development for the treatment of cannabis use disorder and other human diseases.


Assuntos
Canabinoides , Cannabis , Alucinógenos , Animais , Agonistas de Receptores de Canabinoides/farmacologia , Canabinoides/farmacologia , Canabinoides/uso terapêutico , Dronabinol/farmacologia , Humanos , Receptores de Canabinoides
9.
J Neurosci ; 42(11): 2327-2343, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35091501

RESUMO

It is well established that glutamate plays an important role in drug-induced and cue-induced reinstatement of drug seeking. However, the role of glutamate in drug reward is unclear. In this study, we systemically evaluated the effects of multiple glutamate transporter (GLT) inhibitors on extracellular glutamate and dopamine (DA) in the nucleus accumbens (NAc), intravenous cocaine self-administration, intracranial brain-stimulation reward (BSR), and reinstatement of cocaine seeking in male and female rats. Among the five GLT inhibitors we tested, TFB-TBOA was the most potent. Microinjections of TFB-TBOA into the NAc, but not the ventral tegmental area (VTA), or dorsal striatum (DS), dose-dependently inhibited cocaine self-administration under fixed-ratio and progressive-ratio (PR) reinforcement schedules, shifted the cocaine dose-response curve downward, and inhibited intracranial BSR. Selective downregulation of astrocytic GLT-1 expression in the NAc by GLT-1 antisense oligonucleotides also inhibited cocaine self-administration. The reduction in cocaine self-administration following TFB-TBOA administration was NMDA GluN2B receptor dependent, and rats self-administering cocaine showed upregulation of GluN2B expression in NAc DA- and cAMP-regulated phosphoprotein 32 (DARPP-32)-positive medium-spiny neurons (MSNs). In contrast, TFB-TBOA, when locally administered into the NAc, VTA, or ventral pallidum (VP), dose-dependently reinstated cocaine-seeking behavior. Intra-NAc TFB-TBOA-evoked drug-seeking was long-lasting and NMDA/AMPA receptor dependent. These findings, for the first time, indicate that glutamate in the NAc negatively regulates cocaine's rewarding effects, while an excess of glutamate in multiple brain regions can trigger reinstatement of drug-seeking behavior.SIGNIFICANCE STATEMENT It is well known that glutamate plays an important role in relapse to drug seeking. However, the role of glutamate in drug reward is less clear. Here, we report that TFB-TBOA, a highly potent glutamate transporter (GLT) inhibitor, dose-dependently elevates extracellular glutamate and inhibits cocaine self-administration and brain-stimulation reward (BSR), when administered locally into the nucleus accumbens (NAc), but not other brain regions. Mechanistic assays indicate that cocaine self-administration upregulates NMDA-GluN2B receptor subtype expression in striatal dopaminoceptive neurons and activation of GluN2B by TFB-TBOA-enhanced glutamate inhibits cocaine self-administration. TFB-TBOA also reinstates cocaine-seeking behavior when administered into the NAc, ventral tegmental area (VTA), and ventral pallidum (VP). These findings demonstrate that glutamate differentially regulates cocaine reward versus relapse, reducing cocaine reward, while potentiating relapse to cocaine seeking.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Animais , Astrócitos/metabolismo , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Masculino , N-Metilaspartato/farmacologia , Núcleo Accumbens , Ratos , Receptores de N-Metil-D-Aspartato , Autoadministração
11.
Front Pharmacol ; 12: 722476, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34566647

RESUMO

Recent research indicates that brain cannabinoid CB2 receptors are involved in drug reward and addiction. However, it is unclear whether ß-caryophyllene (BCP), a natural product with a CB2 receptor agonist profile, has therapeutic effects on methamphetamine (METH) abuse and dependence. In this study, we used animal models of self-administration, electrical brain-stimulation reward (BSR) and in vivo microdialysis to explore the effects of BCP on METH-taking and METH-seeking behavior. We found that systemic administration of BCP dose-dependently inhibited METH self-administration under both fixed-ratio and progressive-ratio reinforcement schedules in rats, indicating that BCP reduces METH reward, METH intake, and incentive motivation to seek and take METH. The attenuating effects of BCP were partially blocked by AM 630, a selective CB2 receptor antagonist. Genetic deletion of CB2 receptors in CB2-knockout (CB2-KO) mice also blocked low dose BCP-induced reduction in METH self-administration, suggesting possible involvement of a CB2 receptor mechanism. However, at high doses, BCP produced a reduction in METH self-administration in CB2-KO mice in a manner similar as in WT mice, suggesting that non-CB2 receptor mechanisms underlie high dose BCP-produced effects. In addition, BCP dose-dependently attenuated METH-enhanced electrical BSR and inhibited METH-primed and cue-induced reinstatement of drug-seeking in rats. In vivo microdialysis assays indicated that BCP alone did not produce a significant reduction in extracellular dopamine (DA) in the nucleus accumbens (NAc), while BCP pretreatment significantly reduced METH-induced increases in extracellular NAc DA in a dose-dependent manner, suggesting a DA-dependent mechanism involved in BCP action. Together, the present findings suggest that BCP might be a promising therapeutic candidate for the treatment of METH use disorder.

12.
Behav Brain Res ; 415: 113506, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34352292

RESUMO

Recent preclinical studies have reported that pretreatment with the novel and highly-selective dopamine D3 receptor (D3R) antagonists R-VK4-40 or VK4-116 attenuates the abuse-related behavioral effects of oxycodone while enhancing its analgesic properties. However, whether these observed effects are generalizable to the broad class of D3R antagonists and/or extend to opioids other than oxycodone has not been extensively explored. The present study sought to assess the impact of pretreatment with another selective D3R antagonist, PG01037, on several behavioral effects of morphine in mice. C57Bl/6 J mice were pretreated with PG01037 (0-10 mg/kg) and tested for 1) hyperlocomotion induced by acute morphine (5.6-56 mg/kg), 2) locomotor sensitization following repeated morphine (56 mg/kg), 3) antinociception following acute morphine (18 mg/kg), and 4) catalepsy following administration of PG01037 alone or in combination with morphine (56 mg/kg). PG01037 dose-dependently attenuated morphine-induced hyperlocomotion and morphine-induced antinociception at doses that did not alter basal locomotion or nociception alone, but did not prevent the induction of locomotor sensitization following repeated morphine administration. Moreover, PG01037 did not induce catalepsy either alone or in combination with morphine. These results suggest that attenuation of acute opioid-induced hyperactivity may be a behavioral effect shared among D3R-selective antagonists, thus supporting continued investigations into their use as potential treatments for opioid use disorder. However, PG01037 is unlike newer, highly-selective D3R antagonists in its capacity to reduce opioid-induced antinociception, indicating that modulation of opioid analgesia may vary across different D3R antagonists.


Assuntos
Acatisia Induzida por Medicamentos/tratamento farmacológico , Benzamidas/farmacologia , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Entorpecentes/farmacologia , Nociceptividade/efeitos dos fármacos , Piridinas/farmacologia , Receptores de Dopamina D3/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Benzamidas/administração & dosagem , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Piridinas/administração & dosagem
13.
Neuropharmacology ; 189: 108538, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33789118

RESUMO

Cannabinoids produce a number of central nervous system effects via the CB2 receptor (CB2R), including analgesia, antianxiety, anti-reward, hypoactivity and attenuation of opioid-induced respiratory depression. However, the cellular distributions of the CB2Rs in the brain remain unclear. We have reported that CB2Rs are expressed in midbrain dopamine (DA) neurons and functionally regulate DA-mediated behavior(s). Unexpectedly, high densities of CB2-like signaling were also found in a neighboring motor structure - the red nucleus (RN) of the midbrain. In the present study, we systematically explored CB2R expression and function in the RN. Immunohistochemistry and in situ hybridization assays showed high densities of CB2R-immunostaining and mRNA signal in RN magnocellular glutamate neurons in wildtype and CB1-knockout, but not CB2-knockout, mice. Ex vivo electrophysiological recordings in midbrain slices demonstrated that CB2R activation by JWH133 dose-dependently inhibited firing rates of RN magnocellular neurons in wildtype, but not CB2-knockout, mice, while having no effect on RN GABA neurons in transgenic GAD67-GFP reporter mice, suggesting CB2-mediated effects on glutamatergic neurons. In addition, microinjection of JWH133 into the RN produced robust ipsilateral rotations in wildtype, but not CB2-knockout mice, which was blocked by pretreatment with either a CB2 or DA D1 or D2 receptor antagonist, suggesting a DA-dependent effect. Finally, fluorescent tract tracing revealed glutamatergic projections from the RN to multiple brain areas including the ventral tegmental area, thalamus, and cerebellum. These findings suggest that CB2Rs in RN glutamate neurons functionally modulate motor activity, and therefore, constitute a new target in cannabis-based medication development for motor disorders.


Assuntos
Ácido Glutâmico/metabolismo , Atividade Motora/fisiologia , Neurônios/metabolismo , Receptor CB2 de Canabinoide/biossíntese , Núcleo Rubro/metabolismo , Animais , Canabinoides/administração & dosagem , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microinjeções , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/genética , Núcleo Rubro/diagnóstico por imagem
14.
Addict Biol ; 26(4): e13005, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33538103

RESUMO

Despite extensive research, the rewarding effects of cannabinoids are still debated. Here, we used a newly established animal procedure called optogenetic intracranial self-stimulation (ICSS) (oICSS) to re-examine the abuse potential of cannabinoids in mice. A specific adeno-associated viral vector carrying a channelrhodopsin gene was microinjected into the ventral tegmental area (VTA) to express light-sensitive channelrhodopsin in dopamine (DA) neurons of transgenic dopamine transporter (DAT)-Cre mice. Optogenetic stimulation of VTA DA neurons was highly reinforcing and produced a classical "sigmoidal"-shaped stimulation-response curve dependent upon the laser pulse frequency. Systemic administration of cocaine dose-dependently enhanced oICSS and shifted stimulation-response curves upward, in a way similar to previously observed effects of cocaine on electrical ICSS. In contrast, Δ9 -tetrahydrocannabinol (Δ9 -THC), but not cannabidiol, dose-dependently decreased oICSS responding and shifted oICSS curves downward. WIN55,212-2 and ACEA, two synthetic cannabinoids often used in laboratory settings, also produced dose-dependent reductions in oICSS. We then examined several new synthetic cannabinoids, which are used recreationally. XLR-11 produced a cocaine-like increase, AM-2201 produced a Δ9 -THC-like reduction, while 5F-AMB had no effect on oICSS responding. Immunohistochemistry and RNAscope in situ hybridization assays indicated that CB1 Rs are expressed mainly in VTA GABA and glutamate neurons, while CB2 Rs are expressed mainly in VTA DA neurons. Together, these findings suggest that most cannabinoids are not reward enhancing, but rather reward attenuating or aversive in mice. Activation of CB1 R and/or CB2 R in different populations of neurons in the brain may underlie the observed actions.


Assuntos
Canabinoides/efeitos adversos , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Optogenética/métodos , Animais , Comportamento Animal , Cocaína/farmacologia , Neurônios Dopaminérgicos , Dronabinol/farmacologia , Integrases , Masculino , Camundongos , Camundongos Transgênicos , Recompensa , Autoestimulação/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos
15.
Eur Neuropsychopharmacol ; 43: 38-51, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33334652

RESUMO

Cannabinoids produce both rewarding and aversive effects in humans and experimental animals. However, the mechanisms underlying these conflicting findings are unclear. Here we examined the potential involvement of CB1 and CB2 receptors in cannabinoid action using transgenic CB1-knockout (CB1-KO) and CB2-knockout (CB2-KO) mice. We found that Δ9-tetrahydrocannabinol (Δ9-THC) induced conditioned place preference at a low dose (1 mg/kg) in WT mice that was attenuated by deletion of the CB1 receptor. At 5 mg/kg, no subjective effects of Δ9-THC were detected in WT mice, but CB1-KO mice exhibited a trend towards place aversion and CB2-KO mice developed significant place preferences. This data suggests that activation of the CB1 receptor is rewarding, while CB2R activation is aversive. We then examined the nucleus accumbens (NAc) dopamine (DA) response to Δ9-THC using in vivo microdialysis. Unexpectedly, Δ9-THC produced a dose-dependent decrease in extracellular DA in WT mice, that was potentiated in CB1-KO mice. However, in CB2-KO mice Δ9-THC produced a dose-dependent increase in extracellular DA, suggesting that activation of the CB2R inhibits DA release in the NAc. In contrast, Δ9-THC, when administered systemically or locally into the NAc, failed to alter extracellular DA in rats. Lastly, we examined the locomotor response to Δ9-THC. Both CB1 and CB2 receptor mechanisms were shown to underlie Δ9-THC-induced hypolocomotion. These findings indicate that Δ9-THC's variable subjective effects reflect differential activation of cannabinoid receptors. Specifically, the opposing actions of CB1 and CB2 receptors regulate cannabis reward and aversion, with CB2-mediated effects predominant in mice.


Assuntos
Canabinoides , Cannabis , Animais , Dronabinol/farmacologia , Camundongos , Camundongos Knockout , Ratos , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/genética , Receptores de Canabinoides , Recompensa
16.
Curr Opin Pharmacol ; 56: 13-21, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32927246

RESUMO

Pharmacotherapeutics for treatment of psychostimulant use disorder are still an unmet medical goal. Recently, off label use of modafinil (MOD), an approved medication for treatment of sleep disturbances, has been tested as a therapeutic for cocaine and methamphetamine use disorder. Positive results have been found in subjects dependent on psychostimulants without concurrent abuse of other substances. Novel structural analogs of MOD have been synthesized in the search for compounds with potentially broader therapeutic efficacy than the parent drug. In the present report we review their potential efficacy as treatments for psychostimulant abuse and dependence assessed in preclinical tests. Results from these preclinical proof of concept studies reveal that some modafinil analogs do not possess typical cocaine-like neurochemical and behavioral effects. Further, they might blunt the reinforcing effects of psychostimulants in animal models, suggesting their potential efficacy as pharmacotherapeutics for treatment of psychostimulant use disorders.


Assuntos
Estimulantes do Sistema Nervoso Central , Cocaína , Animais , Compostos Benzidrílicos/uso terapêutico , Estimulantes do Sistema Nervoso Central/efeitos adversos , Inibidores da Captação de Dopamina , Humanos , Modafinila
17.
Physiol Behav ; 227: 113164, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32891609

RESUMO

Adolescents represent a large demographic of marijuana consumers. Regrettably, use during this developmental period has been associated with above average health risks. A growing body of evidence suggests that adolescent drug use in the lifetime of a parent can modify behavior and neurochemistry in descendants without direct exposure. The current study was designed to evaluate the effects of pre-conception THC during adolescence on vulnerability to cocaine in adult male offspring. Male and female rats were given an intermittent THC (0 or 1.5 mg/kg) exposure regimen during the adolescent window and mated with drug group conspecifics in adulthood. F1-THC and F1-Veh pups were cross fostered to drug naïve control dams. In Experiment 1, adult offspring underwent cocaine (0 or 15 mg/kg) locomotor sensitization procedures and showed no effect of parental THC exposure on locomotor activity. In Experiment 2, intravenous catheters were implanted and subjects were tested under a number of reinforcement schedules with cocaine (FR1, FR5, FR10, PR, dose-response, extinction, cue + stress induced reinstatement). F1-THC subjects exhibited a slight decrease in cocaine responding during acquisition and a more rapid extinction, but they failed to produce significant differences on any other measure. These findings indicate that adolescent cannabis use likely has minimal effects on cocaine abuse liability in the next generation.


Assuntos
Transtornos Relacionados ao Uso de Cocaína , Cocaína , Animais , Condicionamento Operante , Relação Dose-Resposta a Droga , Dronabinol/farmacologia , Dronabinol/uso terapêutico , Feminino , Masculino , Ratos , Recompensa , Autoadministração
18.
Pharmacol Biochem Behav ; 197: 173001, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32710886

RESUMO

Exposure to environmental stimuli in one generation can produce altered behavioral and neurobiological phenotypes in descendants. Recent work has shown that parental exposure to cannabinoids alters the rewarding properties of other abused drugs in the subsequent generation. However, whether preconception Δ9-tetrahydrocannabinol (THC) administration modifies the affective properties of nicotine in offspring is unknown. To address this question, male and female rats (F0) received THC (0 or 1.5 mg/kg) throughout the adolescent window and were bred on PND 65. In Experiment 1, adult F1-THC and F1-Veh progeny (males and females) underwent nicotine locomotor sensitization procedures during which nicotine (0 or 0.4 mg/kg) was administered every other day for five exposures, and locomotor activity was recorded on each exposure followed by a final nicotine challenge. There was no cross-generational effect of THC on nicotine locomotor sensitization, although acute exposure to nicotine produced greater activity in females relative to males independent of THC history. In Experiment 2, adult F1-THC and F1-Veh progeny (males and females) were implanted with jugular catheters and trained to self-administer nicotine (0.03 mg/kg/infusion). Following acquisition, all subjects were allowed to self-administer nicotine on a number of reinforcement schedules, e.g., FR2, FR5 and PR, followed by dose response and extinction procedures. Across all indices, F1-THC and F1-Veh subjects displayed similar IVSA of nicotine with no sex differences. The fact that there was no evidence of cross-generational effects of THC on nicotine suggests that such effects are drug-specific.


Assuntos
Dronabinol/farmacologia , Fertilização/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Psicotrópicos/farmacologia , Recompensa , Animais , Animais Recém-Nascidos , Feminino , Locomoção/efeitos dos fármacos , Masculino , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Gravidez , Ratos , Esquema de Reforço , Autoadministração , Fatores Sexuais
19.
Drug Alcohol Depend ; 212: 107985, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32386920

RESUMO

BACKGROUND: An emerging area of preclinical research has investigated whether drug use in parents prior to conception influences drug responsivity in their offspring. The present work sought to further characterize such effects with cannabis by examining whether a parental THC history modified locomotor sensitization to morphine and self-administration of heroin in adult progeny. METHODS: Male and female Sprague Dawley rats were exposed to eight injections of 0 or 1.5 mg/kg THC during adolescence and bred with subjects from the same dose group. In Experiment 1, adult male and female offspring (F1-THC and F1-Veh) underwent locomotor sensitization procedures with morphine over five trials followed by a 5-day abstinence period and a final morphine challenge. In Experiment 2, subjects were trained to self-administer heroin and tested under a number of conditions (FR1, FR5, FR10, PR, dose response assessment, extinction, cue- + stress-induced reinstatement). RESULTS: Germline THC exposure had no effect on morphine locomotor sensitization. However, F1-THC males displayed a reduced motivation to self-administer heroin relative to F1-Veh males. CONCLUSIONS: The present data indicate that parental THC exposure alters the reinforcing properties of heroin in a sex-specific manner. As such, mild to moderate cannabis use during adolescence may alter heroin abuse liability for males in the subsequent generation, but have limited effects on females.


Assuntos
Analgésicos Opioides/administração & dosagem , Dronabinol/administração & dosagem , Alucinógenos/administração & dosagem , Heroína/administração & dosagem , Reforço Psicológico , Animais , Relação Dose-Resposta a Droga , Feminino , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Morfina/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Autoadministração , Fatores Sexuais
20.
Exp Clin Psychopharmacol ; 28(1): 32-43, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30998057

RESUMO

In preclinical populations, binge consumption of a high-fat diet (HFD) initiated during either adolescence or adulthood increases the intravenous self-administration (IVSA) of cocaine, whereas ad lib HFD consumption initiated during adulthood reduces or fails to influence cocaine intake. From this, it appears that binge exposure is a sufficient condition to increase cocaine IVSA and that such effects occur independent of the exposure period. It is not clear, however, if ad lib exposure would be sufficient to affect the IVSA of cocaine if initiated during adolescence, a developmental period associated with high-risk behavior. To investigate this question, the present experiment evaluated the effects of consumption of a HFD given throughout adolescence and adulthood on cocaine IVSA (0.75 mg/kg/infusion). Specifically, male Sprague-Dawley rats were maintained on either a HFD (n = 24) or chow diet (n = 15) beginning on postnatal day (PND) 21 and as adults underwent cocaine IVSA [Fixed Ratio (FR) 1, FR 5, FR 10, FR 20, Progressive Ratio (PR) and cue- and drug + cue-induced responding] from PNDs 77-126. Under all of these conditions, animals maintained on the HFD displayed higher rates of cocaine IVSA than those given access to chow. The present data demonstrate that under these specific conditions long-term exposure during the risk period of adolescence and extended throughout adulthood is capable of impacting the subsequent likelihood of cocaine self-administration and suggest that diet type and the duration of exposure may be important factors influencing the vulnerability to drug intake. (PsycINFO Database Record (c) 2020 APA, all rights reserved).


Assuntos
Comportamento Animal , Cocaína/administração & dosagem , Dieta Hiperlipídica , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento Alimentar , Administração Intravenosa , Animais , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Autoadministração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...