Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Behav Pharmacol ; 30(5): 422-428, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30480551

RESUMO

Previous preclinical research suggests that L-methamphetamine (L -MA) has potential therapeutic utility to treat psychostimulant abuse. This study examined potential abuse-related and adverse physiological effects of D -MA and L -MA alone and in combination in rats, as these effects had not been previously characterized. Potential abuse-related effects were examined in locomotor sensitization and conditioned place preference paradigms. Body temperature was monitored to assess the physiological effects of these drugs or drug combinations. In the locomotor study, D-MA induced locomotor sensitization to both D-MA and L -MA. L -MA induced locomotor sensitization only to D-MA. Responses to a combination of L-MA and D -MA were not differentially affected by L-MA or D-MA conditioning. In the conditioned place preference study, D-MA and L -MA each induced significant place preference. L -MA did not attenuate D-MA-induced place preference. In the body temperature study, D-MA induced hyperthermia and L -MA induced hypothermia. In combination, L -MA did not affect D-MA-induced hyperthermia. These data suggest that L -MA alone produces less abuse-related and adverse physiological effects than D-MA, but modulates and is modulated by concurrent and subsequent D-MA exposure, which may enhance the abuse liability of both drugs. These findings should be considered when L -MA is proposed for replacement therapy.


Assuntos
Metanfetamina/análogos & derivados , Metanfetamina/farmacologia , Estereoisomerismo , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Febre , Hipotermia , Locomoção/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
2.
Behav Pharmacol ; 30(5): 429-434, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30383551

RESUMO

Pharmacotherapies for fibromyalgia treatment are lacking. This study examined the antinociceptive and antidepressant-like effects of imidazoline I2 receptor (I2R) agonists in a reserpine-induced model of fibromyalgia in rats. Rats were treated for 3 days with vehicle or reserpine. The von Frey filament test was used to assess the antinociceptive effects of I2 receptor agonists, and the forced swim test was used to assess the antidepressant-like effects of these drugs. 2-BFI (3.2-10 mg/kg, intraperitoneally), phenyzoline (17.8-56 mg/kg, intraperitoneally), and CR4056 (3.2-10 mg/kg, intraperitoneally) all dose-dependently produced significant antinociceptive effects, which were attenuated by the I2R antagonist idazoxan. Only CR4056 significantly reduced the immobility time in the forced swim test in both vehicle-treated and reserpine-treated rats. These data suggest that I2R agonists may be useful to treat fibromyalgia-related pain and comorbid depression.


Assuntos
Hiperalgesia/tratamento farmacológico , Idazoxano/farmacologia , Receptores de Imidazolinas/metabolismo , Analgésicos/farmacologia , Animais , Benzofuranos/farmacologia , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Fibromialgia/tratamento farmacológico , Hiperalgesia/induzido quimicamente , Idazoxano/metabolismo , Imidazóis/farmacologia , Receptores de Imidazolinas/agonistas , Imidazolinas/metabolismo , Imidazolinas/farmacologia , Masculino , Dor/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Reserpina/farmacologia
3.
J Neurosci ; 37(4): 882-892, 2017 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-28123023

RESUMO

A novel G-protein coupled receptor, trace amine-associated receptor 1 (TAAR1), has been shown to be a promising target to prevent stimulant relapse. Our recent studies showed that systemic administration of TAAR1 agonists decreased abuse-related behaviors of cocaine. However, the role of TAAR1 in specific subregions of the reward system in drug addiction is unknown. Here, using a local pharmacological activation method, we assessed the role of TAAR1 within the subregions of the mesocorticolimbic system: that is, the VTA, the prelimbic cortex (PrL), and infralimbic cortex of medial prefrontal cortex, the core and shell of NAc, BLA, and CeA, on cue- and drug-induced cocaine-seeking in the rat cocaine reinstatement model. We first showed that TAAR1 mRNA was expressed throughout these brain regions. Rats underwent cocaine self-administration, followed by extinction training. RO5166017 (1.5 or 5.0 µg/side) or vehicle was microinjected into each brain region immediately before cue- and drug-induced reinstatement of cocaine-seeking. The results showed that microinjection of RO5166017 into the VTA and PrL decreased both cue- and drug priming-induced cocaine-seeking. Microinjection of RO5166017 into the NAc core and shell inhibited cue- and drug-induced cocaine-seeking, respectively. Locomotor activity or food reinforced operant responding was unaffected by microinjection of RO5166017 into these brain regions. Cocaine-seeking behaviors were not affected by RO5166017 when microinjected into the substantia nigra, infralimbic cortex, BLA, and CeA. Together, these results indicate that TAAR1 in different subregions of the mesocorticolimbic system distinctly contributes to cue- and drug-induced reinstatement of cocaine-seeking behavior. SIGNIFICANCE STATEMENT: TAAR1 has been indicated as a modulator of the dopaminergic system. Previous research showed that systemic administration of TAAR1 agonists could attenuate cocaine-related behaviors, suggesting that TAAR1 may be a promising drug target for the treatment of cocaine addiction. However, the specific role of TAAR1 in subregions of the mesocorticolimbic system in drug addiction is unknown. Here, we first showed that TAAR1 mRNA is expressed throughout the subregions of the mesocorticolimbic system. Then, by using a local pharmacological activation method, we demonstrated that TAAR1 in different subregions of the mesocorticolimbic system distinctly contributes to cue- and drug-induced reinstatement of cocaine-seeking behavior.


Assuntos
Tonsila do Cerebelo/fisiologia , Cocaína/administração & dosagem , Neurônios Dopaminérgicos/fisiologia , Comportamento de Procura de Droga/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Dopamina/fisiologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/fisiologia , Masculino , Microinjeções , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Oxazóis/administração & dosagem , Fenetilaminas/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Autoadministração , Área Tegmentar Ventral/fisiologia
4.
Eur J Neurosci ; 47(9): 1087-1095, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29514408

RESUMO

Chronic pain is a significant public health problem with a lack of safe and effective analgesics. The imidazoline I2 receptor (I2 R) is a promising analgesic target, but the neuroanatomical structures involved in mediating I2 R-associated behaviors are unknown. I2 Rs are enriched in the arcuate nucleus, dorsal raphe (DR), interpeduncular nucleus, lateral mammillary body, medial habenula, nucleus accumbens (NAc) and paraventricular nucleus; thus, this study investigated the antinociceptive and hypothermic effects of microinjections of the I2 R agonist 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI). In rats, intra-DR microinjections produced antinociception in complete Freund's adjuvant- and chronic constriction injury-induced pain models. Intra-NAc microinjections produced antinociception and increased noxious stimulus-associated side time in a place escape/avoidance paradigm. Intra-NAc pretreatment with the I2 R antagonist idazoxan but not the D1 receptor antagonist SCH23390 or the D2 receptor antagonist raclopride attenuated intra-NAc 2-BFI-induced antinociception. Intra-NAc idazoxan did not attenuate systemically administered 2-BFI-induced antinociception. Microinjections into the other regions did not produce antinociception, and in none of the regions produced hypothermia. These data suggest that I2 R activation in some but not all I2 R-enriched brain regions is sufficient to produce antinociception and supports the theory that different I2 R-associated effects are mediated via distinct receptor populations, which may in turn be distributed differentially throughout the CNS.


Assuntos
Encéfalo/efeitos dos fármacos , Imidazóis/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Receptores de Dopamina D1/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Benzofuranos/farmacologia , Feminino , Imidazolinas/farmacologia , Masculino , Medição da Dor/métodos , Ratos Sprague-Dawley
5.
Behav Pharmacol ; 28(7): 590-593, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28704278

RESUMO

Increasing evidence suggests that trace amine-associated receptor 1 (TAAR1) is an important modulator of the dopaminergic system. Existing molecular evidence indicates that TAAR1 regulates dopamine levels through interactions with dopamine transporters and D2 receptors. However, investigations to date have not been exhaustive and other pathways may be involved. In this study, we used a well-described set of behaviors, quinpirole-induced yawning and hypothermia, to explore the potential interaction of TAAR1 and D3 receptors, which are members of the 'D2-like' dopamine receptor subfamily. Previous studies have shown that for D2/D3 receptor agonists, the induction of yawning is a D3 receptor-mediated effect, whereas the inhibition of yawning and induction of hypothermia are D2 receptor-mediated effects. Quinpirole produced an inverted U-shaped dose-effect curve for yawning, which was shifted downward dose-dependently by each of the TAAR1 agonists RO5263397 and RO5166017. Quinpirole also produced dose-dependent hypothermia, which was not affected by either TAAR1 agonist. These results suggest that TAAR1 agonists may interact with D3 receptors and/or its downstream pathways, as opposed to D2 receptors. These findings may shed light on a previously unexplored possibility for the mechanism of TAAR1-mediated effects.


Assuntos
Oxazóis/farmacologia , Fenetilaminas/farmacologia , Bocejo/efeitos dos fármacos , Animais , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Hipotermia/induzido quimicamente , Masculino , Quimpirol/metabolismo , Quimpirol/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/agonistas , Receptores de Dopamina D3/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo
6.
J Pharmacol Exp Ther ; 357(3): 509-19, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27056847

RESUMO

Although µ-opioids have been reported to interact favorably with imidazoline I2 receptor (I2R) ligands in animal models of chronic pain, the dependence on the µ-opioid receptor ligand efficacy on these interactions had not been previously investigated. This study systematically examined the interactions between the selective I2 receptor ligand 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI) and three µ-opioid receptor ligands of varying efficacies: fentanyl (high efficacy), buprenorphine (medium-low efficacy), and 17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6α-[(3'-isoquinolyl) acetamido] morphine (NAQ; very low efficacy). The von Frey test of mechanical nociception and Hargreaves test of thermal nociception were used to examine the antihyperalgesic effects of drug combinations in complete Freund's adjuvant-induced inflammatory pain in rats. Food-reinforced schedule-controlled responding was used to examine the rate-suppressing effects of each drug combination. Dose-addition and isobolographical analyses were used to characterize the nature of drug-drug interactions in each assay. 2-BFI and fentanyl fully reversed both mechanical and thermal nociception, whereas buprenorphine significantly reversed thermal but only slightly reversed mechanical nociception. NAQ was ineffective in both nociception assays. When studied in combination with fentanyl, NAQ acted as a competitive antagonist (apparent pA2 value: 6.19). 2-BFI/fentanyl mixtures produced additive to infra-additive analgesic interactions, 2-BFI/buprenorphine mixtures produced supra-additive to infra-additive interactions, and 2-BFI/NAQ mixtures produced supra-additive to additive interactions in the nociception assays. The effects of all combinations on schedule-controlled responding were generally additive. Results consistent with these were found in experiments using female rats. These findings indicate that lower-efficacy µ-opioid receptor agonists may interact more favorably with I2R ligands than high-efficacy µ-opioid receptor agonists.


Assuntos
Analgésicos Opioides/farmacologia , Benzofuranos/farmacologia , Imidazóis/farmacologia , Receptores de Imidazolinas/agonistas , Receptores Opioides mu/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Masculino , Ratos , Ratos Sprague-Dawley
7.
Psychopharmacology (Berl) ; 241(3): 479-487, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38159161

RESUMO

RATIONALE: Increasing evidence shows that imidazoline I2 receptor agonists enhance opioid-induced analgesia, suggesting that the combination of I2 receptor agonists with opioids could be a favorable strategy for pain control. However, the effect of I2 receptor agonists on the abuse liability of opioids is unknown. This study examined the impact of the I2 receptor agonist 2-BFI on some abuse-related behavioral effects of the opioid morphine in rats. OBJECTIVES: The von Frey filament test was used to determine the antinociceptive effects of 2-BFI (intravenous, i.v.) in a rat model of complete Freund's adjuvant (CFA)-induced inflammatory pain. IV self-administration was used to assess the reinforcing effects of 2-BFI alone and to assess the effects of non-contingent injections of 2-BFI (i.p.) on morphine self-administration. A two-lever drug discrimination paradigm in which rats were trained to discriminate 3.2 mg/kg morphine (i.p.) from saline was used to examine whether 2-BFI or another I2 receptor agonist 2-(4,5-dihydroimidazol-2-yl)quinoline hydrochloride (BU224) affected the discriminative stimulus effects of morphine. RESULTS: 2-BFI could not maintain reliable self-administration behavior in rats with no pain or CFA-treated inflammatory pain. However, pretreatment with 2-BFI (i.p.) produced dose-dependent decreases in the dose-effect curve of morphine self-administration. Both 2-BFI and BU224 did not substitute for morphine but significantly attenuated the discriminative stimulus effects of morphine. CONCLUSIONS: These results suggest that I2 receptor agonists do not enhance, but in fact appear to decrease, the abuse liability of opioids, further supporting the potential utility of I2 receptor agonist-opioid combination therapy for pain control.


Assuntos
Benzofuranos , Imidazóis , Imidazolinas , Morfina , Ratos , Animais , Morfina/farmacologia , Morfina/uso terapêutico , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Ratos Sprague-Dawley , Dor/tratamento farmacológico , Relação Dose-Resposta a Droga , Receptores de Imidazolinas/agonistas
8.
Curr Biol ; 31(17): 3797-3809.e5, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34273280

RESUMO

While energy balance is critical to survival, many factors influence food intake beyond caloric need or "hunger." Despite this, some neurons that drive feeding in mice are routinely referred to as "hunger neurons," whereas others are not. To understand how specific hypothalamic circuits control interoceptive hunger, we trained mice to discriminate fasted from sated periods. We then manipulated three hypothalamic neuronal populations with well-known effects on feeding while mice performed this task. While activation of ARCAGRP neurons in sated mice caused mice to report being food-restricted, LHVGAT neuron activation or LHVGLUT2 neuron inhibition did not. In contrast, LHVGAT neuron inhibition or LHVGLUT2 neuron activation in fasted mice attenuated natural hunger, whereas ARCAGRP neuron inhibition did not. Each neuronal population evoked distinct effects on food consumption and reward. After satiety- or sickness-induced devaluation, ARCAGRP neurons drove calorie-specific feeding, while LHVGAT neurons drove calorie-indiscriminate food intake. Our data support a role for ARCAGRP neurons in homeostatic feeding and implicate them in driving a hunger-like internal state that directs behavior toward caloric food sources. Moreover, manipulations of LH circuits did not evoke hunger-like effects in sated mice, suggesting that they may govern feeding more related to reward, compulsion, or generalized consumption than to energy balance, but also that these LH circuits can be powerful negative appetite modulators in fasted mice. This study highlights the complexity of hypothalamic feeding regulation and can be used as a framework to characterize how other neuronal circuits affect hunger and identify potential therapeutic targets for eating disorders.


Assuntos
Fome , Hipotálamo , Proteína Relacionada com Agouti/metabolismo , Animais , Apetite , Ingestão de Alimentos/fisiologia , Fome/fisiologia , Hipotálamo/metabolismo , Camundongos , Neurônios/fisiologia
9.
J Neurosci Methods ; 348: 109015, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33259847

RESUMO

Imaging neuronal activity in awake, behaving animals has become a groundbreaking method in neuroscience that has rapidly enhanced our understanding of how the brain works. In vivo microendoscopic imaging has enabled researchers to see inside the brains of experimental animals and thus has emerged as a technology fit to answer many experimental questions. By combining microendoscopy with cutting edge targeting strategies and sophisticated analysis tools, neuronal activity patterns that underlie changes in behavior and physiology can be identified. However, new users may find it challenging to understand the techniques and to leverage this technology to best suit their needs. Here we present a background and overview of the necessary components for performing in vivo optical calcium imaging and offer some detailed guidance for current recommended approaches.


Assuntos
Encéfalo , Neurônios , Animais , Encéfalo/diagnóstico por imagem , Cálcio , Microscopia de Fluorescência , Neuroimagem
10.
Cell Rep ; 36(8): 109615, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34433027

RESUMO

Assigning behavioral roles to genetically defined neurons within the lateral hypothalamus (LH) is an ongoing challenge. We demonstrate that a subpopulation of LH GABAergic neurons expressing leptin receptors (LHLEPR) specifically drives appetitive behaviors in mice. Ablation of LH GABAergic neurons (LHVGAT) decreases weight gain and food intake, whereas LHLEPR ablation does not. Appetitive learning in a Pavlovian conditioning paradigm is delayed in LHVGAT-ablated mice but prevented entirely in LHLEPR-ablated mice. Both LHVGAT and LHLEPR neurons bidirectionally modulate reward-related behaviors, but only LHVGAT neurons affect feeding. In the Pavlovian paradigm, only LHLEPR activity discriminates between conditioned cues. Optogenetic activation or inhibition of either population in this task disrupts discrimination. However, manipulations of LHLEPR→VTA projections evoke divergent effects on responding. Unlike food-oriented learning, chemogenetic inhibition of LHLEPR neurons does not alter cocaine-conditioned place preference but attenuates cocaine sensitization. Thus, LHLEPR neurons may specifically regulate appetitive behaviors toward non-drug reinforcers.


Assuntos
Comportamento Apetitivo/fisiologia , Comportamento Animal/fisiologia , Região Hipotalâmica Lateral/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Aprendizagem/fisiologia , Camundongos Transgênicos , Optogenética/métodos , Recompensa
11.
Elife ; 102021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-34042586

RESUMO

Understanding how neuronal circuits control nociceptive processing will advance the search for novel analgesics. We use functional imaging to demonstrate that lateral hypothalamic parvalbumin-positive (LHPV) glutamatergic neurons respond to acute thermal stimuli and a persistent inflammatory irritant. Moreover, their chemogenetic modulation alters both pain-related behavioral adaptations and the unpleasantness of a noxious stimulus. In two models of persistent pain, optogenetic activation of LHPV neurons or their ventrolateral periaqueductal gray area (vlPAG) axonal projections attenuates nociception, and neuroanatomical tracing reveals that LHPV neurons preferentially target glutamatergic over GABAergic neurons in the vlPAG. By contrast, LHPV projections to the lateral habenula regulate aversion but not nociception. Finally, we find that LHPV activation evokes additive to synergistic antinociceptive interactions with morphine and restores morphine antinociception following the development of morphine tolerance. Our findings identify LHPV neurons as a lateral hypothalamic cell type involved in nociception and demonstrate their potential as a target for analgesia.


Assuntos
Comportamento Animal , Região Hipotalâmica Lateral/fisiopatologia , Nociceptividade , Dor/fisiopatologia , Dor/psicologia , Analgésicos Opioides/uso terapêutico , Animais , Animais Geneticamente Modificados , Comportamento Animal/efeitos dos fármacos , Sinalização do Cálcio , Modelos Animais de Doenças , Tolerância a Medicamentos , Feminino , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Morfina/farmacologia , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Técnicas de Rastreamento Neuroanatômico , Nociceptividade/efeitos dos fármacos , Optogenética , Dor/metabolismo , Dor/prevenção & controle , Parvalbuminas/genética , Parvalbuminas/metabolismo
12.
Physiol Behav ; 221: 112912, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32289319

RESUMO

Throughout the central nervous system, neurons expressing the calcium-binding protein parvalbumin have been typically classified as GABAergic with fast-spiking characteristics. However, new methods that allow systematic characterization of the cytoarchitectural organization, connectivity, activity patterns, neurotransmitter nature, and function of genetically-distinct cell types have revealed populations of parvalbumin-positive neurons that are glutamatergic. Remarkably, such findings challenge longstanding concepts that fast-spiking neurons are exclusively GABAergic, suggesting conservation of the fast-spiking phenotype across at least two neurotransmitter systems. This review focuses on the recent advancements that have begun to reveal the functional roles of lateral hypothalamic parvalbumin-positive neurons in regulating behaviors essential for survival.


Assuntos
Região Hipotalâmica Lateral , Parvalbuminas , Proteínas de Ligação ao Cálcio , Fenômenos Eletrofisiológicos , Região Hipotalâmica Lateral/metabolismo , Neurônios/metabolismo , Parvalbuminas/metabolismo
13.
Sci Rep ; 9(1): 12026, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31427712

RESUMO

A pivotal role of the lateral hypothalamus (LH) in regulating appetitive and reward-related behaviors has been evident for decades. However, the contributions of LH circuits to other survival behaviors have been less explored. Here we examine how lateral hypothalamic neurons that express the calcium-binding protein parvalbumin (PVALB; LHPV neurons), a small cluster of neurons within the LH glutamatergic circuitry, modulate nociception in mice. We find that photostimulation of LHPV neurons suppresses nociception to an acute, noxious thermal stimulus, whereas photoinhibition potentiates thermal nociception. Moreover, we demonstrate that LHPV axons form functional excitatory synapses on neurons in the ventrolateral periaqueductal gray (vlPAG), and photostimulation of these axons mediates antinociception to both thermal and chemical visceral noxious stimuli. Interestingly, this antinociceptive effect appears to occur independently of opioidergic mechanisms, as antagonism of µ-opioid receptors with systemically-administered naltrexone does not abolish the antinociception evoked by activation of this LHPV→vlPAG pathway. This study directly implicates LHPV neurons in modulating nociception, thus expanding the repertoire of survival behaviors regulated by LH circuits.


Assuntos
Região Hipotalâmica Lateral/fisiologia , Neurônios/metabolismo , Nociceptividade , Parvalbuminas/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Animais , Fenômenos Eletrofisiológicos , Feminino , Masculino , Camundongos , Vias Neurais , Sinapses/fisiologia , Transmissão Sináptica
14.
PLoS One ; 14(7): e0219522, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31291348

RESUMO

Across species, motivated states such as food-seeking and consumption are essential for survival. The lateral hypothalamus (LH) is known to play a fundamental role in regulating feeding and reward-related behaviors. However, the contributions of neuronal subpopulations in the LH have not been thoroughly identified. Here we examine how lateral hypothalamic leptin receptor-expressing (LHLEPR) neurons, a subset of GABAergic cells, regulate motivation in mice. We find that LHLEPR neuronal activation significantly increases progressive ratio (PR) performance, while inhibition decreases responding. Moreover, we mapped LHLEPR axonal projections and demonstrated that they target the ventral tegmental area (VTA), form functional inhibitory synapses with non-dopaminergic VTA neurons, and their activation promotes motivation for food. Finally, we find that LHLEPR neurons also regulate motivation to obtain water, suggesting that they may play a generalized role in motivation. Together, these results identify LHLEPR neurons as modulators within a hypothalamic-ventral tegmental circuit that gates motivation.


Assuntos
Região Hipotalâmica Lateral/fisiologia , Motivação/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Condicionamento Operante/fisiologia , Comportamento Alimentar/psicologia , Feminino , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Modelos Animais , Vias Neurais/fisiologia , Neurônios/fisiologia , Recompensa , Técnicas Estereotáxicas , Sinapses , Área Tegmentar Ventral/citologia
15.
Nat Commun ; 10(1): 4140, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515501

RESUMO

Persistent transcriptional and morphological events in the nucleus accumbens (NAc) and other brain reward regions contribute to the long-lasting behavioral adaptations that characterize drug addiction. Opiate exposure reduces the density of dendritic spines on medium spiny neurons of the NAc; however, the underlying transcriptional and cellular events mediating this remain unknown. We show that heroin self-administration negatively regulates the actin-binding protein drebrin in the NAc. Using virus-mediated gene transfer, we show that drebrin overexpression in the NAc is sufficient to decrease drug seeking and increase dendritic spine density, whereas drebrin knockdown potentiates these effects. We demonstrate that drebrin is transcriptionally repressed by the histone modifier HDAC2, which is relieved by pharmacological inhibition of histone deacetylases. Importantly, we demonstrate that heroin-induced adaptations occur only in the D1+ subset of medium spiny neurons. These findings establish an essential role for drebrin, and upstream transcriptional regulator HDAC2, in opiate-induced plasticity in the NAc.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Neuropeptídeos/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Heroína/efeitos adversos , Histona Desacetilase 2/metabolismo , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/genética , Núcleo Accumbens/metabolismo , Alcaloides Opiáceos/efeitos adversos , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Dor/metabolismo , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
16.
Br J Pharmacol ; 175(9): 1519-1534, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29451703

RESUMO

BACKGROUND AND PURPOSE: Although the antinociceptive efficacies of imidazoline I2 receptor agonists have been established, the exact post-receptor mechanisms remain unknown. This study tested the hypothesis that monoaminergic transmission is critical for I2 receptor agonist-induced antinociception. EXPERIMENTAL APPROACH: von Frey filaments were used to assess antinociceptive effects of two I2 receptor agonists, 2-BFI and CR4056 on chronic constriction injury (CCI)-induced neuropathic pain or complete Freund's adjuvant (CFA)-induced inflammatory pain in rats. Rectal temperature was measured to assess hypothermic effects of 2-BFI. A two-lever drug discrimination paradigm in which rats were trained to discriminate 5.6 mg·kg-1 2-BFI (i.p.) from its vehicle was used to examine the discriminative stimulus effects of 2-BFI. In each experiment, pharmacological mechanisms were investigated by combining 2-BFI or CR4056 with various pharmacological manipulations of the monoaminergic system including selective reuptake inhibition, monoamine depletion and monoamine receptor antagonism. KEY RESULTS: In the CCI model, selective reuptake inhibitors of 5-HT (fluoxetine) or noradrenaline (desipramine), but not dopamine (GBR12909), enhanced 2-BFI-induced antinociception. Selective depletion of 5-HT or noradrenaline almost abolished 2-BFI-induced antinociception. 5-HT1A , 5-HT2A and α1 -adrenoceptor antagonists, but not other monoaminergic antagonists, attenuated 2-BFI and CR4056-induced antinociception in CCI and/or CFA models. However, none of these monoamine receptor antagonists significantly altered 2-BFI-induced hypothermia or discriminative stimulus effects. CONCLUSIONS AND IMPLICATIONS: Antinociception induced by I2 receptor agonists was mediated by serotonergic and noradrenergic mechanisms with 5-HT1A , 5-HT2A and α1 -adrenoceptor being particularly important. In contrast, the hypothermic and discriminative stimulus effects of I2 receptor agonists were mediated by distinct, independent mechanisms.


Assuntos
Analgésicos/farmacologia , Monoaminas Biogênicas/metabolismo , Imidazóis/farmacologia , Receptores de Imidazolinas/agonistas , Quinazolinas/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Discriminação Psicológica/efeitos dos fármacos , Interações Medicamentosas , Hipotermia Induzida , Masculino , Inibidores da Captação de Neurotransmissores/farmacologia , Medição da Dor/efeitos dos fármacos , Ratos , Antagonistas da Serotonina/farmacologia
17.
Neuropharmacology ; 129: 36-46, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29128305

RESUMO

Impulsivity is an important personality trait associated with several clinical syndromes including drug abuse. While repeated drug exposure is known to increase certain behavioral responses, such as locomotion, to subsequent drug exposure, few studies have examined whether such sensitization develops for impulsive behavior. In the current study we tested the effects of methamphetamine acutely, during the course of, and upon discontinuation of chronic methamphetamine treatment on impulsive behavior in two models, the 5-choice serial reaction time task (5-CSRTT) and the delay-discounting task which measure impulsive action and impulsive choice, respectively. We also examined whether the trace amine-associated receptor 1 (TAAR1) agonist RO5263397 attenuated methamphetamine-induced effects in parallel tests. Acute methamphetamine dose-dependently increased premature responses in the 5-CSRTT and shifted the delay function upward in delay discounting. Up to 40 days of methamphetamine treatment did not significantly alter the dose-effect curve of methamphetamine-induced premature responses, but produced a significant effect in the delay-discounting task. RO5263397 attenuated acute methamphetamine-induced premature responses, but this effect became non-significant over the course of chronic treatment. RO5263397 did not significantly alter the delay-discounting performance. Discontinuation of methamphetamine treatment increased premature responses, which was attenuated by RO5263397, but did not significantly alter the delay discounting function. These results suggest that acute discontinuation from prolonged methamphetamine treatment increases impulsivity, which can be reduced by a TAAR1 agonist.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Comportamento Impulsivo/efeitos dos fármacos , Metanfetamina/toxicidade , Oxazóis/uso terapêutico , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/etiologia , Animais , Comportamento de Escolha/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Locomoção/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Tempo
18.
Neuropsychopharmacology ; 43(12): 2435-2444, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29472642

RESUMO

Nicotine addiction and abuse remains a global health issue. To date, the fundamental neurobiological mechanism of nicotine addiction remains incompletely understood. Trace amine-associated receptor 1 (TAAR1) is thought to directly modulate dopaminergic system and are thought to be a neural substrate underlying addictive-like behaviors. We aimed to investigate the role of TAAR1 in nicotine addictive-like behaviors. TAAR1 expression after nicotine treatment was evaluated by western blotting. c-Fos immunofluorescence and in vivo fast-scan cyclic voltammetry were used to examine the activation of brain regions and dopamine release, respectively. We then thoroughly and systematically examined the role of TAAR1 in mediating nicotine-induced sensitization, nicotine discrimination, nicotine self-administration, nicotine demand curve, and the reinstatement of nicotine-seeking. Local pharmacological manipulation was conducted to determine the role of TAAR1 in the nucleus accumbens (NAcs) in the reinstatement of nicotine-seeking. We found that the expression of TAAR1 protein was selectively downregulated in the NAc, with no change in either dorsal striatum or prefrontal cortex. TAAR1 activation was sufficient to block nicotine-induced c-Fos expression in the NAc, while also reducing nicotine-induced dopamine release in the NAc. Systemic administration of TAAR1 agonists attenuated the expression and development of nicotine-induced sensitization, nicotine self-administration, the reinstatement of nicotine-seeking, and increased the elasticity of nicotine demand curve, while intra-NAc infusions of a TAAR1 agonist was sufficient to attenuate nicotine reinstatement. Moreover, TAAR1-knockout rats showed augmented cue-induced and drug-induced reinstatement of nicotine-seeking. These results indicated that modulation of TAAR1 activity regulates nicotine addictive-like behaviors and TAAR1 represents a novel target towards the treatment of nicotine addiction.


Assuntos
Condicionamento Operante/fisiologia , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Núcleo Accumbens/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiologia , Animais , Condicionamento Operante/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Masculino , Núcleo Accumbens/química , Núcleo Accumbens/efeitos dos fármacos , Oxazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Autoadministração
19.
Biochem Pharmacol ; 153: 260-268, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29366977

RESUMO

Chronic pain is a large, unmet public health problem. Recent studies have demonstrated the importance of neuroinflammation in the establishment and maintenance of chronic pain. However, pharmacotherapies that reduce neuroinflammation have not been successfully developed to treat chronic pain thus far. Several preclinical studies have established imidazoline I2 receptor (I2R) agonists as novel candidates for chronic pain therapies, and while some I2R ligands appear to modulate neuroinflammation in certain scenarios, whether they exert anti-neuroinflammatory effects in models of chronic pain is unknown. This study examined the effects of the prototypical I2R agonist 2-(2-benzofuranyl)-2-imidazoline hydrochloride (2-BFI) on hypersensitivity and neuroinflammation induced by chronic constriction injury (CCI), a neuropathic pain model in rats. In CCI rats, twice-daily treatment with 10 mg/kg 2-BFI for seven days consistently increased mechanical and thermal nociception thresholds, reduced GFAP and Iba-1 levels in the dorsal horn of the spinal cord, and reduced levels of TNF-α relative to saline treatment. These results were recapitulated in primary mouse cortical astrocyte cultures. Incubation with 2-BFI attenuated GFAP expression and supernatant TNF-α levels in LPS-stimulated cultures. These results suggest that I2R agonists such as 2-BFI may reduce neuroinflammation which may partially account for their antinociceptive effects.


Assuntos
Benzofuranos/uso terapêutico , Modelos Animais de Doenças , Hiperalgesia/tratamento farmacológico , Imidazóis/uso terapêutico , Receptores de Imidazolinas/agonistas , Neuralgia/tratamento farmacológico , Corno Dorsal da Medula Espinal/efeitos dos fármacos , Animais , Benzofuranos/farmacologia , Células Cultivadas , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Imidazóis/farmacologia , Receptores de Imidazolinas/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Ratos , Ratos Sprague-Dawley , Corno Dorsal da Medula Espinal/metabolismo , Corno Dorsal da Medula Espinal/patologia
20.
Psychopharmacology (Berl) ; 234(22): 3299-3307, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28825118

RESUMO

RATIONALE: Recent research has established the imidazoline I2 receptor as a promising target for the development of novel analgesics. However, despite an increasing understanding of imidazoline I2 receptor-mediated behavioral effects, little is known about post-I2-receptor signaling mechanisms. OBJECTIVE: This study examined the effects of several inhibitors of Ca2+ signaling mechanisms on two behavioral effects of the prototypical imidazoline I2 receptor ligand 2-(2-benzofuranyl)-2-imidazoline (2-BFI). METHODS: The von Frey filament test was used to examine the antinociceptive effects of 2-BFI in complete Freund's adjuvant (CFA)-induced inflammatory pain in rats. A two-lever drug discrimination paradigm in which rats were trained to discriminate 5.6 mg/kg (intraperitoneally) 2-BFI from its vehicle was used to examine the discriminative stimulus effects of 2-BFI. RESULTS: The L-type Ca2+ channel blockers verapamil and nimodipine, the calmodulin antagonist W-7, and the internal Ca2+ release inhibitor ryanodine all attenuated the antinociceptive effects of 2-BFI. Oxycodone- and acetaminophen-induced antinociception was unaffected by pretreatment with the Ca2+ channel blockers. Rats learned to reliably discriminate 5.6 mg/kg 2-BFI from saline. The I2 receptor agonists BU224, RS45041, tracizoline, and CR4056 all fully substituted for 5.6 mg/kg 2-BFI while idazoxan, S22687, 2,5-dimethoxy-4-methylamphetamine (DOM), and phenyzoline produced partial or no substitution. Verapamil, nimodipine, and W-7 did not alter the discriminative stimulus effects of 2-BFI. CONCLUSION: These results indicate that the antinociceptive effects of 2-BFI involve intracellular Ca2+ elevation and/or downstream Ca2+/calmodulin signaling, whereas the discriminative stimulus effects of 2-BFI are mediated by a distinct, independent mechanism.


Assuntos
Analgésicos/farmacologia , Benzofuranos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Aprendizagem por Discriminação/efeitos dos fármacos , Imidazóis/farmacologia , Receptores de Imidazolinas/agonistas , Analgésicos/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Masculino , Limiar da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA