Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Zhen Ci Yan Jiu ; 49(5): 441-447, 2024 May 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38764114

RESUMO

OBJECTIVES: To observe the effect of electroacupuncture (EA) at "Neiguan" (PC6) on pain response in mice injected with complete Freund's adjuvant (CFA) in the hind paw, so as to investigate the mechanism of orexin 1 receptor (OX1R) -endogenous cannabinoid 1 receptor (CB1R) pathway in acupuncture analgesia. METHODS: A total of 48 male C57BL/6 mice were used in the present study. In the first part of this study, 18 mice were randomized into control, model and EA groups, with 6 mice in each group. In the second part of this study, 30 mice were randomized into control, model, EA, EA+Naloxone, EA+OX1R antagonist (SB33486) groups, with 6 mice in each group. Inflammatory pain model was established by subcutaneous injection of 20 µL CFA solution in the left hind paw. EA (2 Hz, 2 mA ) was applied to bilateral PC6 for 20 min, once a day for 5 consecutive days. The mice in the EA+Naloxone and EA+SB33486 groups were intraperitoneally injected with naloxone (10 mg/kg) or SB33486 (15 mg/kg) 15 min before EA intervention on day 5, respectively. Tail-flick method and Von Frey method were used to detect the thermal pain threshold and mechanical pain threshold of mice. Quantitative real-time PCR was used to detect the expression level of ß-endorphin mRNA in periaqueductal gray (PAG) of mice. The expression of OX1R positive cells in the lateral hypothalamic area (LH) and CB1R positive cells in the ventrolateral periaqueductal gray (vlPAG) were detected by immunofluorescence. RESULTS: Compared with the control group, the thermal pain threshold and mechanical pain threshold of the model group were decreased (P<0.001), the expression level of ß-endorphin mRNA in PAG was decreased (P<0.001), and the numbers of OX1R positive cells in LH and CB1R positive cells in vlPAG were decreased (P<0.05, P<0.001). Compared with the model group, the thermal pain threshold and mechanical pain threshold of the EA group were significantly increased (P<0.001), and the numbers of OX1R positive cells in LH and CB1R positive cells in vlPAG were increased (P<0.01, P<0.001). Compared with the EA group, the mechanical pain threshold in the EA+SB33486 group was significantly decreased (P<0.01), but there was no significant difference in the mechanical pain threshold between the EA+Naloxone group and EA group, and the numbers of OX1R positive neurons in LH and CB1R positive neurons in vlPAG were decreased in the EA+SB33486 group (P<0.001). CONCLUSIONS: EA at PC6 can achieve analgesic effect on CFA mice by activating the OX1R-CB1R pathway in the brain, and this effect is opioid-independent.


Assuntos
Pontos de Acupuntura , Encéfalo , Eletroacupuntura , Receptores de Orexina , Dor , Animais , Humanos , Masculino , Camundongos , Encéfalo/metabolismo , Inflamação/terapia , Inflamação/metabolismo , Inflamação/genética , Camundongos Endogâmicos C57BL , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Dor/metabolismo , Dor/genética , Manejo da Dor
2.
Int J Eat Disord ; 57(7): 1418-1432, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38456603

RESUMO

OBJECTIVE: Test the efficacy of the selective orexin 1 receptor (OX1R) antagonist (SO1RA) nivasorexant in an animal model of binge-eating disorder (BED) and study its dose-response relationship considering free brain concentrations and calculated OX1R occupancy. Compare nivasorexant's profile to that of other, structurally diverse SO1RAs. Gain understanding of potential changes in orexin-A (OXA) neuropeptide and deltaFosB (ΔFosB) protein expression possibly underlying the development of the binge-eating phenotype in the rat model used. METHOD: Binge-like eating of highly palatable food (HPF) in rats was induced through priming by intermittent, repeated periods of dieting and access to HPF, followed by an additional challenge with acute stress. Effects of nivasorexant were compared to the SO1RAs ACT-335827 and IDOR-1104-2408. OXA expression in neurons and neuronal fibers as well as ΔFosB and OXA-ΔFosB co-expression was studied in relevant brain regions using immuno- or immunofluorescent histochemistry. RESULTS: All SO1RAs dose-dependently reduced binge-like eating with effect sizes comparable to the positive control topiramate, at unbound drug concentrations selectively blocking brain OX1Rs. Nivasorexant's efficacy was maintained upon chronic dosing and under conditions involving more frequent stress exposure. Priming for binge-like eating or nivasorexant treatment resulted in only minor changes in OXA or ΔFosB expression in few brain areas. DISCUSSION: Selective OX1R blockade reduced binge-like eating in rats. Neither ΔFosB nor OXA expression proved to be a useful classifier for their binge-eating phenotype. The current results formed the basis for a clinical phase II trial in BED, in which nivasorexant was unfortunately not efficacious compared with placebo. PUBLIC SIGNIFICANCE: Nivasorexant is a new investigational drug for the treatment of binge-eating disorder (BED). It underwent clinical testing in a phase II proof of concept trial in humans but was not efficacious compared with placebo. The current manuscript investigated the drug's efficacy in reducing binge-like eating behavior of a highly palatable sweet and fat diet in a rat model of BED, which initially laid the foundation for the clinical trial.


Assuntos
Transtorno da Compulsão Alimentar , Modelos Animais de Doenças , Antagonistas dos Receptores de Orexina , Receptores de Orexina , Animais , Antagonistas dos Receptores de Orexina/farmacologia , Ratos , Masculino , Transtorno da Compulsão Alimentar/tratamento farmacológico , Receptores de Orexina/metabolismo , Ratos Sprague-Dawley , Orexinas/metabolismo
3.
J Psychopharmacol ; 38(3): 305-308, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38327032

RESUMO

In rodents, orexin neuropeptides regulate motivation and reward-seeking via orexin 1 receptor (OX1R) signaling in the mesolimbic dopaminergic system. This role is clearly established for rewards inherent to drugs of abuse but less so for natural rewards. Reported effects of the selective OX1R antagonist (SO1RA) SB-334867 on motivation for palatable food are ambiguous. In our experimental conditions neither SB-334867, nor two additional, structurally different SO1RAs, ACT-335827 and the clinical development candidate nivasorexant, affected effort-based responding for sucrose in rats. The positive control lisdexamfetamine, approved for psychiatric disorders associated with altered reward sensitivity such as binge eating disorder, increased effort-based responding.


Assuntos
Benzoxazóis , Naftiridinas , Recompensa , Sacarose , Ureia/análogos & derivados , Humanos , Ratos , Animais , Orexinas/farmacologia , Receptores de Orexina , Sacarose/farmacologia , Condicionamento Operante
4.
IBRO Neurosci Rep ; 15: 386-394, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38074161

RESUMO

The hippocampal-prefrontal cortex network dynamics is reported to be involved in various cognitive functions and in different mood disturbances including depression. It has been suggested that blocking orexin-1 receptors can be beneficial in depression. The purpose of this study is to determine whether orexin-1 receptor antagonists have an impact on changes in brain oscillations in the hippocampus and prefrontal cortex in a rat model of depression. Forty-eight male Wistar rats were divided into six experimental groups: control, chronic mild stress (CMS), acute SB-334867, a selective orexin-1 receptor antagonist, treated rats (SB), chronic SB-treated (CSB), CMS+SB, and CMS+CSB. Two stainless steel recording electrodes were placed in the coordinates of the hippocampus (HPC) and the prefrontal cortex (PFC). After behavioral verification of the model, local field potentials were recorded at 1 kHz sampling frequency. The absolute power of different frequency bands was obtained using the Fast Fourier Transform (FFT) function, and the power spectral density (PSD) of each frequency band was calculated for each animal. In the CMS- treated animals, the low-gamma band power increased both in the HPC and PFC (p ≤ 0.05), which were reversed by chronic SB-334867 treatment (p ≤ 0.05). The alterations in theta, and high-gamma band power were not significant in CMS treated rats, while acute and chronic SB-334867 treatment diminished the theta and high-gamma band power (p ≤ 0.05), respectively. The hippocampal-prefrontal coherence decreased in the delta (p ≤ 0.01), theta (p ≤ 0.01), and alpha (p ≤ 0.05) band range of the CMS exposed rats. It is concluded that CMS boosts the low-gamma band power, which is reversed by CSB treatment. The low-frequency band coherence is attenuated after CMS treatment.

5.
Pharmacol Res Perspect ; 11(5): e01143, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37800597

RESUMO

Nivasorexant, a selective orexin-1-receptor antagonist, has recently been assessed in the treatment of humans with binge-eating disorder. Herein, the inhibitory potential of nivasorexant on cytochromes P450 (CYPs) 2C9, 2C19, and 3A4 was evaluated. Human liver microsomes/recombinant CYP enzymes were evaluated in vitro. In vivo, a single-center, open-label, fixed-sequence study was performed in healthy adults to explore the effect of 100 mg nivasorexant administered twice daily (b.i.d.) on the pharmacokinetics (PK) of flurbiprofen (50 mg, CYP2C9), omeprazole (20 mg, CYP2C19), midazolam (2 mg, CYP3A4) making use of a cocktail approach. Plasma PK sampling was performed over 24 h on Day 1 (Cocktail alone), 8 (Cocktail + nivasorexant), and 15 (Cocktail + nivasorexant at steady state). Genotyping of subjects' CYPs was performed while safety and tolerability were also assessed. In vitro, nivasorexant inhibited CYP2C9, 2C19, and 3A4 in competitive inhibition assays with IC50 values of 8.6, 1.6, and 19-44 µM, respectively, while showing a significant time-dependent CYP2C19 inhibition. In 22 subjects, exposure to flurbiprofen, omeprazole, and midazolam was generally higher during concomitant single- (i.e., area under the plasma concentration-time curve [AUC] ratio increased by 1.04-, 2.05-, and 1.56-fold, respectively) and repeated-dose (i.e., AUC ratio increased by 1.47-, 6.84-, and 3.71-fold, respectively) nivasorexant administration compared with the cocktail substrates administered alone. The most frequently reported adverse event was somnolence. According to regulatory guidance, nivasorexant is classified as a moderate CYP2C19 and weak CYP3A4 inhibitor after 1 day and as a weak CYP2C9, strong CYP2C19, and moderate CYP3A4 inhibitor after 8 days of 100 mg b.i.d. administration. Clinicaltrials.gov ID: NCT05254548.


Assuntos
Flurbiprofeno , Midazolam , Adulto , Humanos , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2C9 , Orexinas , Inibidores do Citocromo P-450 CYP3A , Voluntários Saudáveis , Interações Medicamentosas , Sistema Enzimático do Citocromo P-450/genética , Omeprazol/farmacocinética
6.
Int J Eat Disord ; 56(11): 2120-2130, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37584285

RESUMO

OBJECTIVE: This Phase II, placebo-controlled, double-blind study investigated the efficacy, safety, and tolerability of nivasorexant in the treatment of adults with moderate to severe binge-eating disorder (BED). METHODS: Adults meeting the DSM-5 BED criteria were randomized 1:1 to placebo or nivasorexant (100 mg b.i.d.). The primary endpoint was the change from baseline to Week 12 in the number of binge eating (BE) days per week. Exploratory efficacy endpoints included cessation of BE in the last 4 weeks of treatment; and change from baseline to Week 12 in the number of BE episodes/week, the clinician global impression (CGI) of change, the Yale-Brown Obsessive-Compulsive Scale modified for BE, and the Hamilton rating scale for depression (HAMD-17). Key safety outcomes included treatment-emergent adverse events (TEAEs) and adverse events of special interest (i.e., somnolence and fatigue). RESULTS: Sixty-eight participants were randomized to each treatment arm. The change from baseline to Week 12 in the number of BE days/week was the same for placebo (least squares mean [LSM]: -2.93) and nivasorexant (LSM: -2.93), with no difference between the treatment groups (LSM difference = .000 [95% confidence interval (CI): -.69, .69], p = .9992). Furthermore, no differences between treatment groups were observed in the exploratory efficacy endpoints. Nivasorexant was well tolerated; the overall incidence of TEAEs was balanced between treatment groups, and the frequency of somnolence and fatigue in the nivasorexant group were similar to placebo. DISCUSSION: In this proof-of-concept study, 100 mg b.i.d. nivasorexant did not improve BE in adults with moderate to severe BED. PUBLIC SIGNIFICANCE: The results of this Phase II study indicate that nivasorexant was well tolerated in adults with BED, but did not improve binge eating behavior over placebo. Further research is needed to improve our understanding of the role of the orexin-1 receptor in BED.


Assuntos
Transtorno da Compulsão Alimentar , Bulimia , Humanos , Adulto , Transtorno da Compulsão Alimentar/tratamento farmacológico , Transtorno da Compulsão Alimentar/induzido quimicamente , Dimesilato de Lisdexanfetamina/uso terapêutico , Sonolência , Método Duplo-Cego , Resultado do Tratamento
7.
J Psychopharmacol ; 37(6): 577-589, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37165642

RESUMO

BACKGROUND: Up to 40% of patients suffering from anxiety disorders do not benefit from currently available pharmacological treatments. Overactivity of the orexin-1 receptor (OX1R) has been implicated in anxiety- and panic-related states. AIM & METHODS: We investigated the pharmacokinetics and characterized the pharmacodynamic (PD) profile of the OX1R antagonist JNJ-61393215 using a battery of central nervous system assessments investigating relevant functional domains such as alertness, attention, (visuo)motor coordination, balance, subjective effects and resting-state electroencephalography in a single ascending dose placebo-controlled study in doses from 1 to 90 mg inclusive, assessing PD up to 10 h after dosing, safety and pharmacokinetic in 48 healthy male subjects. RESULTS: Average time to maximal plasma concentration (Tmax) ranged between 1.0 and 2.25 h; average half-life ranged from 13.6 to 24.6 h and average maximum plasma concentration ranged from 1.4 to 136.8 ng/mL in the 1 and 90 mg groups, respectively. JNJ-61393215 did not demonstrate any statistically significant or clinically meaningful effects on any PD endpoint at any dose investigated at Tmax nor over the total period up to 10 h post-dose and was well tolerated. The reported somnolence rate was 16.7% (which was attributable to the cohorts receiving 6 mg and higher doses) compared to 12.5% in placebo. CONCLUSION: This observation is in line with our knowledge about the OX1R in preclinical studies, where only inconsistent and non-dose-dependent changes in electroencephalography or other behavioural measures were observed under non-challenged conditions, potentially exemplifying the need for a challenged subject.


Assuntos
Antagonistas dos Receptores de Orexina , Humanos , Masculino , Área Sob a Curva , Relação Dose-Resposta a Droga , Método Duplo-Cego , Meia-Vida , Voluntários Saudáveis , Antagonistas dos Receptores de Orexina/efeitos adversos , Antagonistas dos Receptores de Orexina/farmacocinética , Orexinas
8.
Neuromolecular Med ; 25(2): 286-300, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36689149

RESUMO

Rapid eye movement (REM) sleep behavior disorder (RBD) is a powerful early sign of Parkinson's disease (PD), but the pathogenetic mechanism involved in RBD remains largely unexplored. α-Synuclein has been verified to form Lewy bodies in the orexin neurons, whose activity and function rely on the orexin 1 receptor (OX1R). Dysfunction of the OX1R may induce the occurrence of RBD. Here, we determined the role of the interaction between α-Synuclein and OX1R in the pathogenesis of RBD, in vitro and in vivo. We found that injection of α-Synuclein into the lateral hypothalamus area (LHA) damaged orexin neurons and induced the RBD-like sleep pattern, to further damage dopaminergic neurons and result in locomotor dysfunction in mice. α-Synuclein interacted with OX1R, promoting the degradation of OX1R through proteasomal and lysosomal pathways. In addition, overexpression of α-Synuclein downregulated OX1R-mediated signaling, subsequently leading to orexin neuron damage. We conclude that α-Synuclein induced the occurrence of RBD via interaction with OX1R and modulated its degradation. These findings provide evidence for a novel mechanism by which the association of α-Synuclein with OX1R was attributed to α-Synuclein-induced orexin neuron damage, which may be a new molecular target for an effective therapeutic strategy for RBD pathology.


Assuntos
Doença de Parkinson , Transtorno do Comportamento do Sono REM , Camundongos , Animais , alfa-Sinucleína/metabolismo , Orexinas , Doença de Parkinson/complicações , Transtorno do Comportamento do Sono REM/complicações , Transtorno do Comportamento do Sono REM/diagnóstico , Neurônios Dopaminérgicos/patologia
9.
Behav Brain Res ; 438: 114171, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36280008

RESUMO

Cognitive dysfunction is the main clinical manifestation of Alzheimer's disease (AD). Previous research found that elevated orexin level in the cerebrospinal fluid was closely related to the course of AD, and orexin-A treatment could increase amyloid ß protein (Aß) deposition and aggravate spatial memory impairment in APP/PS1 mice. Furthermore, recent research found that dual orexin receptor (OXR) antagonist might affect Aß level and cognitive dysfunction in AD, but the effects of OX1R or OX2R alone is unreported until now. Considering that OX1R is highly expressed in the hippocampus and plays important roles in learning and memory, the effects of OX1R in AD cognitive dysfunction and its possible mechanism should be investigated. In the present study, selective OX1R antagonist SB-334867 was used to block OX1R. Then, different behavioral tests were performed to observe the effects of OX1R blockade on cognitive function of 3xTg-AD mice exhibited both Aß and tau pathology, in vivo electrophysiological recording and western blot were used to investigate the potential mechanism. The results showed that chronic OX1R blockade aggravated the impairments of short-term working memory, long-term spatial memory and synaptic plasticity in 9-month-old female 3xTg-AD mice, increased levels of soluble Aß oligomers and p-tau, and decreased PSD-95 expression in the hippocampus of 3xTg-AD mice. These results indicate that the detrimental effects of SB-334867 on cognitive behaviors in 3xTg-AD mice are closely related to the decrease of PSD-95 and depression of in vivo long-term potentiation (LTP) caused by increased Aß oligomers and p-tau.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Camundongos , Animais , Feminino , Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Orexinas/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Hipocampo/metabolismo , Antagonistas dos Receptores de Orexina/farmacologia , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo
10.
Basic Clin Neurosci ; 13(3): 407-420, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36457878

RESUMO

Introduction: Introduction: Several studies have demonstrated that orexins may regulate different forms of affective and cognitive processes during wakefulness. The Orbitofrontal Cortex (OFC) and Anterior Cingulate Cortex (ACC), as an essential part of the Prefrontal Cortex (PFC), have a crucial role in cognitive processes such as reward and decision-making. They also have a high amount of orexin receptor type 1 (OX1Rs). Methods: In the present study, we inhibited OX1Rs in this area after a 10-min baseline recording to find out the role of OX1Rs in the OFC neuron's firing rate. Next, we inhibited the lateral hypothalamus (LH) as the primary source of orexinergic neurons. Afterward, using a single-unit recording technique in rats, we detected the effects of the lateral hypothalamus on the firing rate and activity pattern of the ACC or OFC neurons. Results: Data showed that the blockade of OX1Rs in the OFC could excite 8 and inhibit 1 neuron(s) out of 11. In addition, the blockade of OX1Rs in the ACC could excite 6 and inhibit 3 neurons out of 10. LH inactivation excited 5 out of 12 neurons and inhibited 6 in the ACC. It also excited 8 and inhibited 6 neurons out of 14 in the OFC. These data suggest that the blockade of OX1Rs excites 72% of the neurons, but LH inactivation had a stimulating effect on only 50% of neurons in two main subregions of the PFC. Conclusion: Accordingly, PFC neurons may receive the orexinergic inputs from the LH and indirectly from other sources. Highlights: Blockade of orexin receptors 1 in ACC alter neural activity in this region.Blockade of orexin receptors 1 in OFC alter neural activity in this region.Lateral hypothalamus inhibition increase neural activity of only 50% neurons in ACC and OFC. Plain Language Summary: The discovery of the neuropeptide orexin/hypocretin in 1998 and subsequent research during the past 20 years revealed an important role for the lateral hypothalamus (LH) in driving the reward pathway, sleep and awake circuits, decision making and psychotic disorders. But our knowledge towards the function of this neurotransmitter on neural activity in specific areas of the brain is limited. In this study we decided to clear the specific role of orexin receptors placed in the two crucial areas of medial prefrontal cortex (mPFC) and the orexin projections from LH on neural firing rates in those regions. In the present study, we investigated the following items by using an in vivo extracellular single-unit recording technique in rats and our data shown that the effect of blocking of orexin receptors1 in mPFC caused a different results than inhibition of the origin of orexin projection in LH as a source of it.

11.
J Pharmacol Sci ; 150(4): 244-250, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36344046

RESUMO

Orexins are produced in hypothalamic areas and orexin-containing neurons are distributed in widespread areas of the central nervous system. Orexins regulate several physiological functions such as arousal, food intake and autonomic control. The presence of orexin-containing neuron terminals and orexin receptors has been confirmed in the nucleus tractus solitarius (NTS), which receives primary afferent fibers from peripheral organs including baroreceptors. However, the neuronal effects of orexin-1 receptor (OX1R) activation were not examined. Here, we aimed to determine the effects of OX1R activation on excitatory synaptic transmission. OX1R activation increased the frequency of spontaneous excitatory synaptic currents (sEPSCs). This effect was blocked by the prior application of L-NAME. In contrast, the amplitude of evoked excitatory postsynaptic currents (eEPSCs) was suppressed by OX1R activation, and this effect was prevented by a cannabinoid receptor 1 blocker, AM251, but not by the pretreatment with L-NAME. Altogether, these results suggest that OX1R activation increases sEPSCs frequency by stimulating NO production, whereas it inhibits eEPSCs by releasing endocannabinoids in the NTS. Thus, OX1R activation had distinct effects on spontaneous and evoked excitatory synaptic transmissions in the NTS.


Assuntos
Núcleo Solitário , Transmissão Sináptica , Ratos , Animais , Orexinas/farmacologia , Técnicas de Patch-Clamp , NG-Nitroarginina Metil Éster/farmacologia , Potenciais Pós-Sinápticos Excitadores
12.
Physiol Behav ; 257: 114000, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36270509

RESUMO

Binge eating disorder (BED) is the most prevalent eating disorder in the adult population. It is characterized by recurrent episodes of uncontrollable overconsumption of palatable food (PF). BED is connected to several comorbidities such as obesity, major depression, and substance use disorder, and was linked to heightened levels of impulsivity. The neurobiological basis of BED is however still vaguely known. Binge eating (BE) occurs without homeostatic needs, and therefore, relates to hedonic consumption of PF. A major brain structure in the control of hedonic feeding, and part of the network regulating impulsive action, is the nucleus accumbens shell (NAcSh). The present study in rats assessed the influence of trait impulsivity on the development of BE-like feeding and the role of the orexigenic neuropeptide orexin (OX) and the anorexigenic neuropeptide cocaine- and amphetamine-regulated transcript (CART) within the NAcSh in a BE-model. The rats were initially ranked for their trait impulsivity based on a screening in the 5-choice serial reaction time task. They were subsequently introduced into a limited access-model to establish BE-like feeding with pure vegetable fat to examine a correlation between trait impulsivity and the development of BE-like feeding. The effect of bilateral infusions of the OX 1-receptor (OX1R) antagonist SB-334867 (SB) and CART-antibodies (CART-ABs) into the NAcSh was examined in relation to trait impulsivity. Further, accumbal OX1R density was evaluated by immunohistochemical staining in rats with normal and BE-like feeding behavior. We found that all animals developed stable BE-like PF intake, independent of trait impulsivity and without differences in the dynamics. The blockade of accumbal OX1Rs effectively reduced PF intake only in the control group that had daily access to PF, with impulsivity trait as a decisive factor, pointing towards alterations in orexinergic transmission in the NAcSh of rats bingeing on pure fat. This was corroborated by a lower density of OX1Rs in the NAcSh of rats with BE-like feeding behavior, precisely, in low-impulsive bingeing rats. Regardless of impulsivity trait, antagonizing CART in the NAcSh did not affect PF intake of control or bingeing animals. This suggests that endogenous accumbal CART does not influence consummatory behavior in ad libitum-fed rats with access to fat, under both normal and BE-like feeding patterns.


Assuntos
Transtorno da Compulsão Alimentar , Bulimia , Cocaína , Neuropeptídeos , Animais , Ratos , Masculino , Núcleo Accumbens/metabolismo , Orexinas , Comportamento Impulsivo/fisiologia , Neuropeptídeos/metabolismo , Anfetaminas , Ingestão de Alimentos/fisiologia
13.
Pharmacol Biochem Behav ; 219: 173449, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35973584

RESUMO

This study was done to evaluate the effect of co-treatment of orexin agents along with citalopram on the modulation of depression-like behavior and the expression of BDNF in the prefrontal cortex (PFC) of sleep-deprived male mice. A sleep deprivation model was performed in which rapid eye movement (REM) sleep was completely prohibited, and non-REM sleep was intensely reduced for 24 h. For drug microinjection, the guide cannula was surgically fixed in the left lateral ventricle of mice. Furthermore, we used the open-field test (OFT), forced swim test (FST), tail suspension test (TST), and splash test for recording depression-like behavior as well as Real-Time PCR amplification for assessing the expression of BDNF in the PFC of REM sleep-deprived mice. Our results revealed that REM sleep deprivation did not change locomotor activity while increased depressive-like behavior in FST, TST, and splash tests. However, the expression of BDNF was decreased in the PFC. Intraperitoneally (i.p.) administration of citalopram induced antidepressant effect in the normal and REM sleep-deprived mice. Moreover, intracerebroventricular (i.c.v.) microinjection of a non-effective dose of SB-334867, an orexin antagonist, potentiated the antidepressant-like effect of citalopram. On the other hand, a non-significant dosage of orexin-1 reversed the antidepressant effect of citalopram in the normal and REM sleep-deprived animals. Furthermore, our results showed that injection of citalopram alone or with SB-334867 increased the mRNA expression level of BDNF in the PFC of REM sleep-deprived mice. These data suggest that REM sleep deprivation interferes with the neural systems underlying the depression-like process and supports a likely interaction of the orexin system with citalopram on the modulation of depression-like behavior in REM sleep-deprived mice.


Assuntos
Citalopram , Sono REM , Animais , Antidepressivos , Benzoxazóis , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citalopram/farmacologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Masculino , Camundongos , Naftiridinas , Orexinas , Privação do Sono/tratamento farmacológico , Ureia/análogos & derivados
14.
Neuropharmacology ; 215: 109168, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35724928

RESUMO

Fear-associated memories and behavior are often expressed in contexts/environments distinctively different from those in which they are created. This generalization process contributes to psychological disorders, particularly PTSD. Stress-related neurocircuits in the basolateral amygdala (BLA) receive inputs from hypothalamic orexin (Orx) neurons, which mediate neuronal activity by targeting orexin 1 (Orx1R) and orexin 2 (Orx2R) receptors via opposing functions. In BLA, inhibition of Orx1R or activation of Orx2R ameliorate stress responsiveness and behavior. We discovered that most Orx1R+ cells also express CamKIIα, while a majority of Orx2R+ cells are colocalized with GAD67. Further, Orx1R gene Hcrtr1 expression was positively correlated, and Orx2R gene Hcrtr2 expression was negatively correlated, with freezing in a phenotype-dependent fashion (Escape vs Stay) in the Stress Alternatives Model (SAM). The SAM consists of 4-days of social interaction between test mice and novel larger aggressors. Exits positioned at opposite ends of the SAM oval arena provide opportunities to actively avoid aggression. By Day 2, mice commit to behavioral phenotypes: Escape or Stay. Pharmacologically manipulating Orx receptor activity in the BLA, before Day 3 of the SAM, was followed with standard tests of anxiety: Open Field (OF) and Elevated Plus Maze (EPM). In Stay mice, freezing in response to social conflict and locomotion during SAM interaction (not home cage locomotion) were generalized to OF, and blocked by intra-BLA Orx1R antagonism, but not Orx2R antagonism. Moreover, patterns of social avoidance for Escape and Stay mice were recapitulated in OF, with generalization mediated by Orx1R and Orx2R antagonism, plus Orx2R stimulation.


Assuntos
Aprendizagem da Esquiva , Complexo Nuclear Basolateral da Amígdala , Receptores de Orexina , Comportamento Social , Sequência de Aminoácidos , Animais , Aprendizagem da Esquiva/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Camundongos , Receptores de Orexina/fisiologia , Orexinas , Fragmentos de Peptídeos , Tripsina
15.
Front Cell Neurosci ; 16: 812359, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35496914

RESUMO

The orexin system comprises two G protein-coupled receptors, OX1 and OX2 receptors (OX1R and OX2R, respectively), along with two endogenous agonists cleaved from a common precursor (prepro-orexin), orexin-A (OX-A) and orexin-B (OX-B). For the receptors, a complex array of signaling behaviors has been reported. In particular, it becomes obvious that orexin receptor coupling is very diverse and can be tissue-, cell- and context-dependent. Here, the early signal transduction interactions of the orexin receptors will be discussed in depth, with particular emphasis on the direct G protein interactions of each receptor. In doing so, it is evident that ligands, additional receptor-protein interactions and cellular environment all play important roles in the G protein coupling profiles of the orexin receptors. This has potential implications for our understanding of the orexin system's function in vivo in both central and peripheral environments, as well as the development of novel agonists, antagonists and possibly allosteric modulators targeting the orexin system.

16.
Behav Brain Res ; 424: 113791, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35151793

RESUMO

Orexin neuropeptides are well known for their role in sleep/wake cycle, feeding behavior and motivation-related behaviors. However, their role in cognition is not clearly understood. We recently published that orexin deficiency impairs intra-dimensional set shifting in female homozygous orexin-deficient mice but improves the first reversal phase in male homozygous orexin-deficient mice in the attentional set shifting task (ASST), a well-established rodent test for cognitive flexibility. In the present study, we tested if intracerebroventricular injections of the selective orexin 1 receptor antagonist SB-334867 (4 µg/2 µL) affects cognitive flexibility in the different phases of ASST. We found that SB-334867 injections impaired the first and second reversal phases in female C57BL/6J mice but not in males. In addition, we also showed that at this particular dose of SB-334867, the consumption of the reward that was used in the ASST was not affected in both males and females. Our findings indicate that cognitive flexibility is impaired by orexin 1 receptor antagonism in a sex-dependent manner and reiterates the sexually dimorphic role of orexin in cognitive flexibility.


Assuntos
Benzoxazóis , Naftiridinas , Animais , Benzoxazóis/farmacologia , Cognição , Feminino , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Orexina , Orexinas , Ureia/análogos & derivados , Ureia/farmacologia
17.
J Comp Neurol ; 530(3): 592-606, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34387361

RESUMO

Orexin neurons play an important role in stress-related mental disorders including post-traumatic stress disorder (PTSD). Anxiety- and depression-related symptoms commonly occur in combination with PTSD. However, the role of the orexin system in mediating alterations in these affective symptoms remains unclear. The medial prefrontal cortex (mPFC) is implicated in both cognitive and emotional processing. In the present study, we investigated anxiety- and depression-related behavioral changes using the elevated plus maze, the sucrose preference test, and the open field test in male rats with single prolonged stress (SPS) induced-PTSD. The expression of orexin-A in the hypothalamus and orexin receptors (OX1R and OX2R) in the mPFC was detected and quantified by immunohistochemistry, western blotting, and real-time polymerase chain reaction. We found that the SPS rats exhibited enhanced levels of anxiety, reduced exploratory activities, and anhedonia. Furthermore, SPS resulted in reductions in the expression of orexin-A in the hypothalamus and the increased the expression of OX1R in the mPFC. The intracerebroventricular administration of orexin-A alleviated behavioral changes in SPS rats and partly restored the increased levels of OX1R in the mPFC. These results suggest that the orexin system plays a role in the anxiety- and depression-related symptoms observed in PTSD.


Assuntos
Transtornos de Estresse Pós-Traumáticos , Animais , Ansiedade , Depressão/etiologia , Modelos Animais de Doenças , Humanos , Masculino , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar , Transtornos de Estresse Pós-Traumáticos/metabolismo
18.
Nutr Neurosci ; 25(5): 1105-1114, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-33151127

RESUMO

The orexin peptides promote hedonic intake and other reward behaviors through different brain sites. The opioid dynorphin peptides are co-released with orexin peptides but block their effects on reward in the ventral tegmental area (VTA). We previously showed that in the paraventricular hypothalamic nucleus (PVN), dynorphin and not orexin peptides enhance hedonic intake, suggesting they have brain-site-specific effects. Obesity alters the expression of orexin and dynorphin receptors, but whether their expression across different brain sites is important to hedonic intake is unclear. We hypothesized that hedonic intake is regulated by orexin and dynorphin peptides in PVN and that hedonic intake in obesity correlates with expression of their receptors. Here we show that in mice, injection of DYN-A1-13 (an opioid dynorphin peptide) in the PVN enhanced hedonic intake, whereas in the VTA, injection of OXA (orexin-A, an orexin peptide) enhanced hedonic intake. In PVN, OXA blunted the increase in hedonic intake caused by DYN-A1-13. In PVN, injection of norBNI (opioid receptor antagonist) reduced hedonic intake but a subsequent OXA injection failed to increase hedonic intake, suggesting that OXA activity in PVN is not influenced by endogenous opioid activity. In the PVN, DYN-A1-13 increased the intake of the less-preferred food in a two-food choice task. In obese mice fed a cafeteria diet, orexin 1 receptor mRNA across brain sites involved in hedonic intake correlated with fat preference but not caloric intake. Together, these data support that orexin and dynorphin peptides regulate hedonic intake in an opposing manner with brain-site-specific effects.


Assuntos
Dinorfinas , Núcleo Hipotalâmico Paraventricular , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Encéfalo/metabolismo , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Camundongos , Obesidade/metabolismo , Orexinas/metabolismo
19.
Peptides ; 146: 170651, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34560171

RESUMO

Orexin 1 receptors (Orx1R) and cannabinoid 1 receptors (CB1R) are implicated in migraine pathophysiology. This study evaluated the potential involvement of Orx1R and CB1R within the ventrolateral periaqueductal gray matter (vlPAG) in the modulation of anxiety-like behavior and social interaction of migraineurs rats. A rat model of migraine induced by recurrent administration of nitroglycerin (NTG) (5 mg/kg/i.p.). The groups of rats (n = 6) were then subjected to intra-vlPAG microinjection of orexin-A (25, 50 pM), and Orx1R antagonist SB334867 (20, 40 nM) or AM 251 (2, 4 µg) as a CB1R antagonist. Behavioral responses were evaluated in elevated plus maze (EPM), open field (OF) and three-chambered social test apparatus. NTG produced a marked anxiety like behaviors, in both EPM and OF tasks. It did also decrease social performance. NTG-related anxiety and social conflicts were attenuated by orexin-A (25, 50 pM). However, NTG effects were exacerbated by SB334867 (40 nM) and AM251 (2, 4 µg). The orexin-A-mediated suppression of NTG-induced anxiety and social conflicts were prevented by either SB334867 (20 nM) or AM251 (2 µg). The findings suggest roles for Orx1R and CB1R signaling within vlPAG in the modulation of migraine-induced anxiety-like behavior and social dysfunction in rats.


Assuntos
Ansiedade/prevenção & controle , Comportamento Animal , Transtornos de Enxaqueca/complicações , Receptores de Orexina/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Comportamento Social , Animais , Ansiedade/etiologia , Benzoxazóis/farmacologia , Masculino , Naftiridinas/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar , Receptores de Neuropeptídeos/efeitos dos fármacos , Ureia/análogos & derivados , Ureia/farmacologia
20.
Neuropharmacology ; 198: 108764, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34450116

RESUMO

Self-control problems are a typical character of drug addiction and excessive food consumption and it has been shown that natural rewards and drugs of abuse share parts of the same neural substrate and reward processing in the brain. Different brain areas are involved in natural and drug reward processing including the mesolimbic pathway, amygdala, nucleus accumbens (NAc), and prefrontal cortex. Considering the important role of orexins in the addictive behavior and the presence of orexin-1 subtype receptors (Orx1R) in the medial prefrontal cortex (mPFC), this study investigated the role of mPFC in natural- and drug-reward seeking behaviors to deepen our understanding of possible similarities or differences. To induce food- or morphine-conditioned place preference (CPP), adult male Wistar rats underwent CPP testing and received intra-mPFC doses of SB334867 (3, 10, or 30 nM/0.5 µl DMSO 12%), as an Orx1R antagonist, during the acquisition or expression phases of the CPP test. Results indicated that microinjection of Orx1R antagonist into the mPFC had similar effects on both morphine- and food-induced CPP and attenuated CPP scores in the acquisition and expression phases of the CPP test. The data demonstrated that Orx1Rs in the mPFC regulate the reward-related effects of morphine- and food-induced reward.


Assuntos
Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Alimentos , Dependência de Morfina/metabolismo , Morfina/farmacologia , Receptores de Orexina/metabolismo , Animais , Benzoxazóis/farmacologia , Comportamento de Procura de Droga , Masculino , Naftiridinas/farmacologia , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Wistar , Recompensa , Ureia/análogos & derivados , Ureia/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA