Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Sci Rep ; 14(1): 7690, 2024 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-38565870

RESUMO

Tunicates are evolutionary model organisms bridging the gap between vertebrates and invertebrates. A genomic sequence in Ciona intestinalis (CiOX) shows high similarity to vertebrate orexin receptors and protostome allatotropin receptors (ATR). Here, molecular phylogeny suggested that CiOX is divergent from ATRs and human orexin receptors (hOX1/2). However, CiOX appears closer to hOX1/2 than to ATR both in terms of sequence percent identity and in its modelled binding cavity, as suggested by molecular modelling. CiOX was heterologously expressed in a recombinant HEK293 cell system. Human orexins weakly but concentration-dependently activated its Gq signalling (Ca2+ elevation), and the responses were inhibited by the non-selective orexin receptor antagonists TCS 1102 and almorexant, but only weakly by the OX1-selective antagonist SB-334867. Furthermore, the 5-/6-carboxytetramethylrhodamine (TAMRA)-labelled human orexin-A was able to bind to CiOX. Database mining was used to predict a potential endogenous C. intestinalis orexin peptide (Ci-orexin-A). Ci-orexin-A was able to displace TAMRA-orexin-A, but not to induce any calcium response at the CiOX. Consequently, we suggested that the orexin signalling system is conserved in Ciona intestinalis, although the relevant peptide-receptor interaction was not fully elucidated.


Assuntos
Ciona intestinalis , Animais , Humanos , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Orexinas/metabolismo , Ciona intestinalis/genética , Ciona intestinalis/metabolismo , Células HEK293 , Transdução de Sinais , Vertebrados/metabolismo , Proteínas de Transporte/metabolismo
2.
Am J Drug Alcohol Abuse ; 50(1): 84-94, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38295363

RESUMO

Background: Methamphetamine use disorder (MUD) is a worldwide health concern. The hypothalamic orexin system regulates stress response and addictive behaviors. The genetic variation in the hypocretin receptor 2 (HCRTR2), rs2653349, is associated with substance use disorder.Objectives: We explored the gene-environment (GxE) interaction of rs2653349 and adverse childhood experiences (ACEs) associated with MUD susceptibility.Methods: Four hundred and one individuals (336 males, 65 females) with MUD and 348 healthy controls (288 males, 60 females) completed a self-report questionnaire evaluating ACEs, encompassing childhood abuse and household dysfunction categories, and were genotyped for SNP rs2653349. Methamphetamine use variables were collected using the Diagnostic Interview for Genetic Studies. We used regression analyses to assess the GxE effect on MUD risk.Results: The MUD group had a comparable genotypic distribution for rs2653349 to the control group, albeit with a higher prevalence and number of types of ACEs, correlating with an increased MUD risk (p < .05). No significant genetic impact of rs2653349 on MUD risk was found. However, we observed a GxE interaction effect between the minor allele of rs2653349 and the number of childhood abuse or household dysfunction types, correlating with a reduced MUD risk (OR = -0.71, p = .04, Benjamini-Hochberg adjusted p = .08 and OR = -0.59, p = .045, Benjamini-Hochberg adjusted p = .09, respectively).Conclusion: HCRTR2 SNP rs2653349 has no significant impact on MUD risk, but ACEs may increase this risk. GxE results suggest that rs2653349 could offer protection against developing MUD in individuals experiencing multiple types of ACEs.


Assuntos
Experiências Adversas da Infância , Metanfetamina , Masculino , Feminino , Humanos , Criança , Receptores de Orexina/genética , Polimorfismo de Nucleotídeo Único/genética , Metanfetamina/efeitos adversos , Genótipo
3.
Structure ; 32(3): 352-361.e5, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38194963

RESUMO

Orexin neuropeptides have many physiological roles in the sleep-wake cycle, feeding behavior, reward demands, and stress responses by activating cognitive receptors, the orexin receptors (OX1R and OX2R), distributed in the brain. There are only subtle differences between OX1R and OX2R in the orthosteric site, which has hindered the rational development of subtype-selective antagonists. In this study, we utilized solution-state NMR to capture the structural plasticity of OX2R labeled with 13CH3-ε-methionine in complex with antagonists. Mutations in the orthosteric site allosterically affected the intracellular tip of TM6. Ligand exchange experiments with the subtype-selective EMPA and the nonselective suvorexant identified three methionine residues that were substantially perturbed. The NMR spectra suggested that the suvorexant-bound state exhibited more structural plasticity than the EMPA-bound state, which has not been foreseen from the close similarity of their crystal structures, providing insights into dynamic features to be considered in understanding the ligand recognition mode.


Assuntos
Metionina , Humanos , Orexinas , Ligantes , Receptores de Orexina/genética , Receptores de Orexina/química , Espectroscopia de Ressonância Magnética
4.
eNeuro ; 11(2)2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38199807

RESUMO

Orexins, which are produced within neurons of the lateral hypothalamic area, play a pivotal role in the regulation of various behaviors, including sleep/wakefulness, reward behavior, and energy metabolism, via orexin receptor type 1 (OX1R) and type 2 (OX2R). Despite the advanced understanding of orexinergic regulation of behavior at the circuit level, the precise distribution of orexin receptors in the brain remains unknown. Here, we develop a new branched in situ hybridization chain reaction (bHCR) technique to visualize multiple target mRNAs in a semiquantitative manner, combined with immunohistochemistry, which provided comprehensive distribution of orexin receptor mRNA and neuron subtypes expressing orexin receptors in mouse brains. Only a limited number of cells expressing both Ox1r and Ox2r were observed in specific brain regions, such as the dorsal raphe nucleus and ventromedial hypothalamic nucleus. In many brain regions, Ox1r-expressing cells and Ox2r-expressing cells belong to different cell types, such as glutamatergic and GABAergic neurons. Moreover, our findings demonstrated considerable heterogeneity in Ox1r- or Ox2r-expressing populations of serotonergic, dopaminergic, noradrenergic, cholinergic, and histaminergic neurons. The majority of orexin neurons did not express orexin receptors. This study provides valuable insights into the mechanism underlying the physiological and behavioral regulation mediated by the orexin system, as well as the development of therapeutic agents targeting orexin receptors.


Assuntos
Núcleo Dorsal da Rafe , Receptores Acoplados a Proteínas G , Camundongos , Animais , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Núcleo Dorsal da Rafe/metabolismo , Mapeamento Encefálico , Hibridização In Situ , RNA Mensageiro
5.
J Neurol ; 270(10): 5064-5070, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37418013

RESUMO

PURPOSE: Cluster headache (CH) is a debilitating condition with severe and recurrent headaches characterized by circannual and circadian rhythms. A genetic contingent was suggested, and several loci were described in large cohorts. However, no variant associated with CH for multiplex families has been described. The purpose of our study was to examine candidate genes and new genetic variants in a multigenerational family of cluster headaches in which two members have original chronobiological characteristics that we have called the phenomenon of "family periodicity". METHODS AND RESULTS: We performed a whole genome sequencing in four patients in a large multigenerational family of cluster headache to identify additional loci associated with CH. This allowed us to replicate the genomic association of HCRTR2 and CLOCK as candidate genes. In two family members with the same phenotypic circadian pattern (familial periodicity) the association of polymorphism NM_001526.4:c.922G > A was shown in the HCRTR2 gene, and NM_004898.4:c.213T > C in the CLOCK gene. INTERPRETATION: This whole genome sequencing reproduced two genetic risk loci for CH already involved in its pathogenicity. This is the first time that the combination of HCRTR2 and CLOCK gene variants is identified in a multigenerational family of CH with striking periodicity characteristics. Our study supports the hypothesis that the combination of HCRTR2 and CLOCK gene variants can contribute to the risk of cluster headache and offer the prospect of a new area of research on the molecular circadian clock.


Assuntos
Cefaleia Histamínica , Humanos , Ritmo Circadiano/genética , Cefaleia Histamínica/genética , Família , Genótipo , Receptores de Orexina/genética , Periodicidade , Fenótipo
6.
Proc Natl Acad Sci U S A ; 120(20): e2220353120, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37155875

RESUMO

Early-life stress has long-term impacts on the structure and function of the anterior cingulate cortex (ACC), and raises the risk of adult neuropsychiatric disorders including social dysfunction. The underlying neural mechanisms, however, are still uncertain. Here, we show that, in female mice, maternal separation (MS) during the first three postnatal weeks results in social impairment accompanied with hypoactivity in pyramidal neurons (PNs) of the ACC. Activation of ACC PNs ameliorates MS-induced social impairment. Neuropeptide Hcrt, which encodes hypocretin (orexin), is the top down-regulated gene in the ACC of MS females. Activating ACC orexin terminals enhances the activity of ACC PNs and rescues the diminished sociability observed in MS females via an orexin receptor 2 (OxR2)-dependent mechanism. Our results suggest orexin signaling in the ACC is critical in mediating early-life stress-induced social impairment in females.


Assuntos
Neuropeptídeos , Estresse Psicológico , Animais , Feminino , Camundongos , Giro do Cíngulo , Privação Materna , Neuropeptídeos/metabolismo , Receptores de Orexina/genética , Orexinas/genética , Orexinas/metabolismo
7.
Peptides ; 165: 171009, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37054895

RESUMO

Orexin-A and -B (identical to hypocretin-1 and -2) are neuropeptides synthesized in the lateral hypothalamus and perifornical area, and orexin neurons project their axon terminals broadly throughout the entire central nervous system (CNS). The activity of orexins is mediated by two specific G protein-coupled receptors (GPCRs), termed orexin type1 receptor (OX1R) and orexin type2 receptor (OX2R). The orexin system plays a relevant role in various physiological functions, including arousal, feeding, reward, and thermogenesis, and is key to human health. Orexin neurons receive various signals related to environmental, physiological, and emotional stimuli. Previous studies have reported that several neurotransmitters and neuromodulators influence the activation or inhibition of orexin neuron activity. In this review, we summarize the modulating factors of orexin neurons in the sleep/wake rhythm and feeding behavior, particularly in the context of the modulation of appetite, body fluids, and circadian signaling. We also describe the effects of life activity, behavior, and diet on the orexin system. Some studies have observed phenomena that have been verified in animal experiments, revealing the detailed mechanism and neural pathway, while their applications to humans is expected in future research.


Assuntos
Receptores de Orexina , Orexinas , Animais , Humanos , Neuropeptídeos/metabolismo , Neurotransmissores/farmacologia , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sono/fisiologia
8.
J Pharmacol Exp Ther ; 385(3): 193-204, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37001988

RESUMO

Loss of orexin neurons is associated with narcolepsy type 1 (NT1), which is characterized by multiple symptoms including excessive daytime sleepiness and cataplexy. Orexin 2 receptor (OX2R) knockout (KO) mice, but not orexin 1 receptor (OX1R) KO mice, show narcolepsy-like phenotypes, thus OX2R agonists are potentially promising for treating NT1. In fact, in early proof-of-concept studies, intravenous infusion of danavorexton, an OX2R-selective agonist, significantly increased wakefulness in individuals with NT1. However, danavorexton has limited oral availability. Here, we report pharmacological characteristics of a novel OX2R agonist, TAK-994 [N-{(2S,3S)-1-(2-hydroxy-2-methylpropanoyl)-2-[(2,3',5'-trifluorobiphenyl-3-yl)methyl]pyrrolidin-3-yl}methanesulfonamide sesquihydrate]. TAK-994 activated recombinant human OX2R (EC50 value of 19 nM) with > 700-fold selectivity against OX1R and activated OX2R-downstream signaling similar to those by orexin peptides in vitro. Oral administration of TAK-994 promoted wakefulness in normal mice but not in OX2R KO mice. TAK-994 also ameliorated narcolepsy-like symptoms in two mouse models of narcolepsy: orexin/ataxin-3 mice and orexin-tTA;TetO diphtheria toxin A mice. The wake-promoting effects of TAK-994 in orexin/ataxin-3 mice were maintained after chronic dosing for 14 days. These data suggest that overall in vitro and in vivo properties, except oral availability, are very similar between TAK-994 and danavorexton. Preclinical characteristics of TAK-994 shown here, together with upcoming clinical study results, can improve our understanding for orally available OX2R agonists as new therapeutic drugs for NT1 and other hypersomnia disorders. SIGNIFICANCE STATEMENT: Narcolepsy type 1 (NT1) is caused by a loss of orexin neurons, and thus an orexin 2 receptor (OX2R) agonist is considered to address the underlying pathophysiology of NT1. Oral administration of TAK-994, a novel OX2R agonist, promoted wakefulness in normal mice, but not in OX2R knockout mice, and ameliorated fragmentation of wakefulness and cataplexy-like episodes in mouse models of narcolepsy. These findings indicate that TAK-994 is an orally available brain-penetrant OX2R-selective agonist with potential to improve narcolepsy-like symptoms.


Assuntos
Cataplexia , Narcolepsia , Camundongos , Humanos , Animais , Cataplexia/tratamento farmacológico , Vigília , Ataxina-3 , Sono/genética , Narcolepsia/tratamento farmacológico , Narcolepsia/genética , Orexinas/genética , Orexinas/metabolismo , Orexinas/farmacologia , Encéfalo/metabolismo , Camundongos Knockout , Receptores de Orexina/agonistas , Receptores de Orexina/genética , Receptores de Orexina/uso terapêutico
9.
J Am Heart Assoc ; 12(6): e028987, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36892078

RESUMO

Background The hypocretin/orexin system has been shown to play a role in heart failure. Whether it also influences myocardial infarction (MI) outcomes is unknown. We evaluated the effect of the rs7767652 minor allele T associated with decreased transcription of the hypocretin/orexin receptor-2 and circulating orexin A concentrations on mortality risk after MI. Methods and Results Data from a single-center, prospectively designed registry of consecutive patients hospitalized for MI at a large tertiary cardiology center were analyzed. Patients without previous history of MI or heart failure were included. A random population sample was used to compare allele frequencies in the general population. Out of 1009 patients (aged 64±12 years, 74.6% men) after MI, 6.1% were homozygotes (TT) and 39.4% heterozygotes (CT) for minor allele. Allele frequencies in the MI group did not differ from 1953 subjects from general population (χ2 P=0.62). At index hospitalization, MI size was the same, but ventricular fibrillation and the need for cardiopulmonary resuscitation were more prevalent in the TT allele variant. Among patients with ejection fraction ≤40% at discharge, the TT variant was associated with a lower increase in left ventricular ejection fraction during follow-up (P=0.03). During the 27-month follow-up, there was a statistically significant association of the TT variant with increased mortality risk (hazard ratio [HR], 2.83; P=0.001). Higher circulating orexin A was associated with a lower mortality risk (HR, 0.41; P<0.05). Conclusions Attenuation of hypocretin/orexin signaling is associated with increased mortality risk after MI. This effect may be partially explained by the increased arrhythmic risk and the effect on the left ventricular systolic function recovery.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Masculino , Humanos , Feminino , Orexinas/genética , Volume Sistólico , Função Ventricular Esquerda , Receptores de Orexina/genética
10.
Peptides ; 164: 170979, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36841281

RESUMO

The orexins (OXs) were first reported in hypothalamus of rat, and they play an important role in diverse physiological actions. The OXs consist of orexin A (OXA) and orexin B (OXB) peptides and their actions are mediated via two G-protein-coupled receptors, orexin 1 receptor (OX1R) and orexin 2 receptor (OX2R), respectively. Presence of OXA and OX1R has been also reported in peripheral organs like reproductive tissues. These findings, therefore, highlight a possible role of OXs and their receptors in male reproductive health. Though, expression and localization of OXB and OX2R in the testis and their role in spermatogenesis are not finally clarified. Herein, we elucidated the localization and the patterns of expression of OXB and OX2R in Parkes mice testes during postnatal development. Results suggest that the precursor prepro-orexin (PPO), OXB and OX2R are expressed at the transcript and protein levels in mouse testis throughout the postnatal development. Immunostaining further showed the localization of OXB and OX2R both in interstitium and tubular compartments of the testis. On 7 day postpartum (7 dpp), only spermatogonia showed immunoreactivity of OXB and OX2R, while at 14, 28, 42 and 90 dpp, immunolocalization of OXB and OX2R were noted in the seminiferous tubules, especially in leptotene, pachytene spermatocytes, round and elongating spermatids, and in Leydig cells and Sertoli cells. The immunoreactivity of OXB and OX2R appeared to be stage-specific in adult mouse testis. The results suggest the expression of OXB and OX2R in mouse testis and their possible regulatory role in spermatogenesis and steroidogenesis.


Assuntos
Espermátides , Testículo , Animais , Masculino , Camundongos , Células Intersticiais do Testículo/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Orexinas/metabolismo , Espermátides/metabolismo , Testículo/metabolismo
11.
Int J Biol Macromol ; 229: 873-884, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36587646

RESUMO

Pacific abalone (Haliotis discus hannai) is a typical nocturnal organism. To examine the circadian expression pattern of orexin receptor type 2 (OX2R) and its potential effect on the feeding behavior of abalone, the coding region sequence of OX2R that is 1215 bp in length and encodes 404 amino acids was first cloned using the rapid amplification of cDNA ends technique. A recombinant expression vector was constructed for H. discus hannai based on the OX2R protein, obtaining a recombinant protein with a molecular weight of 46 kDa. Polyclonal antibody was prepared with the purified recombinant protein used as the antigen, and the antibody titer of ≥512 K was detected by enzyme-linked immunosorbent assay. The expression levels of OX2R determined using western blotting were highest in the intestinal tract (P < 0.05), but they were not significantly different from the levels in the pedal. Immunofluorescence experiments affirmed that OX2R was widely expressed in the columnar cells of the intestinal mucosal epithelium. To further account for the relationship between the onset of feeding behavior and the expression level of OX2R in abalone, the circadian expression characteristics of OX2R were analyzed by dissecting the intestinal tissues after three days of normal feeding and fasting and following the refeeding treatment. The expression levels of OX2R in the refeeding group were significantly higher than those in the normal feeding and fasting groups at any time point (P < 0.05). The cosine curve analysis revealed that the expression levels of OX2R lost rhythmicity after fasting. Based on the quantification of behavioral data for abalone after fasting and refeeding, the cumulative movement distance and movement duration in each group followed a significant cosine rhythm (P < 0.05), which is consistent with abalone's nocturnal ecological habits. However, the cumulative movement distance and movement duration in the fasting group were significantly lower than those in the normal feeding and refeeding groups (P < 0.05). The peak phases of the cumulative movement distance and movement duration in the refeeding group (ZT08:22 and ZT08:44) shifted backward compared to the normal feeding group (ZT07:33 and ZT07:39). The above results first identified the structural characteristics and circadian expression patterns of OX2R in the marine mollusk abalone, which may reveal the molecular mechanism behind the generation of a feeding rhythm in marine nocturnal organisms and serve as a tool helping to maintain the diversity of marine benthic resources.


Assuntos
Jejum , Gastrópodes , Animais , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Sequência de Bases , Proteínas Recombinantes/genética , DNA Complementar/genética , Gastrópodes/genética
12.
Structure ; 30(12): 1582-1589.e4, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36417909

RESUMO

Orexin receptors are a family of G protein-coupled receptors that consist of two subtypes: orexin-1 receptors (OX1Rs) and OX2Rs. They are expressed throughout the central nervous system and are involved in regulating the sleep-wake cycle. The development of antagonists to orexin receptors has become important in drug discovery because modulation of these receptors can lead to novel treatments for diseases related to the regulation of sleep and wakefulness, such as insomnia. In this study, we determined that the structure of OX2R bound to lemborexant, a dual orexin receptor antagonist (DORA), at 2.89 Å resolution. Comparisons of kinetic and dynamic properties of DORAs based on structures and simulations suggest that the enthalpy of molecular binding to receptors and the entropy derived from intramolecular structure can be separately controlled. These results complement existing structural information and allow us to discuss the usefulness of pharmacophore models and target selectivity to OXRs.


Assuntos
Desenho de Fármacos , Piridinas , Orexinas , Receptores de Orexina/genética
13.
Proc Natl Acad Sci U S A ; 119(35): e2207531119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994639

RESUMO

Narcolepsy type 1 (NT1) is a sleep disorder caused by a loss of orexinergic neurons. Narcolepsy type 2 (NT2) is heterogeneous; affected individuals typically have normal orexin levels. Following evaluation in mice, the effects of the orexin 2 receptor (OX2R)-selective agonist danavorexton were evaluated in single- and multiple-rising-dose studies in healthy adults, and in individuals with NT1 and NT2. In orexin/ataxin-3 narcolepsy mice, danavorexton reduced sleep/wakefulness fragmentation and cataplexy-like episodes during the active phase. In humans, danavorexton administered intravenously was well tolerated and was associated with marked improvements in sleep latency in both NT1 and NT2. In individuals with NT1, danavorexton dose-dependently increased sleep latency in the Maintenance of Wakefulness Test, up to the ceiling effect of 40 min, in both the single- and multiple-rising-dose studies. These findings indicate that OX2Rs remain functional despite long-term orexin loss in NT1. OX2R-selective agonists are a promising treatment for both NT1 and NT2.


Assuntos
Estimulantes do Sistema Nervoso Central , Narcolepsia , Receptores de Orexina , Adulto , Animais , Ataxina-3/genética , Ataxina-3/metabolismo , Cataplexia/tratamento farmacológico , Cataplexia/genética , Estimulantes do Sistema Nervoso Central/farmacologia , Estimulantes do Sistema Nervoso Central/uso terapêutico , Modelos Animais de Doenças , Humanos , Camundongos , Narcolepsia/tratamento farmacológico , Narcolepsia/genética , Neurônios/metabolismo , Receptores de Orexina/agonistas , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Receptores de Orexina/uso terapêutico , Orexinas/genética , Orexinas/metabolismo , Fenótipo , Vigília/efeitos dos fármacos , Vigília/genética
14.
PLoS One ; 17(7): e0271901, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35867683

RESUMO

Acquired loss of hypothalamic orexin (hypocretin)-producing neurons causes the chronic sleep disorder narcolepsy-cataplexy. Orexin replacement therapy using orexin receptor agonists is expected as a mechanistic treatment for narcolepsy. Orexins act on two receptor subtypes, OX1R and OX2R, the latter being more strongly implicated in sleep/wake regulation. However, it has been unclear whether the activation of only OX2R, or both OX1R and OX2R, is required to replace the endogenous orexin functions in the brain. In the present study, we examined whether the selective activation of OX2R is sufficient to rescue the phenotype of cataplexy and sleep/wake fragmentation in orexin knockout mice. Intracerebroventricular [Ala11, D-Leu15]-orexin-B, a peptidic OX2R-selective agonist, selectively activated OX2R-expressing histaminergic neurons in vivo, whereas intracerebroventricular orexin-A, an OX1R/OX2R non-selective agonist, additionally activated OX1R-positive noradrenergic neurons in vivo. Administration of [Ala11, D-Leu15]-orexin-B extended wake time, reduced state transition frequency between wake and NREM sleep, and reduced the number of cataplexy-like episodes, to the same degree as compared with orexin-A. Furthermore, intracerebroventricular orexin-A but not [Ala11, D-Leu15]-orexin-B induced drug-seeking behaviors in a dose-dependent manner in wild-type mice, suggesting that OX2R-selective agonism has a lower propensity for reinforcing/drug-seeking effects. Collectively, these findings provide a proof-of-concept for safer mechanistic treatment of narcolepsy-cataplexy through OX2R-selective agonism.


Assuntos
Cataplexia , Narcolepsia , Animais , Cataplexia/tratamento farmacológico , Modelos Animais de Doenças , Comportamento de Procura de Droga , Camundongos , Camundongos Knockout , Narcolepsia/tratamento farmacológico , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/farmacologia , Sono/fisiologia , Vigília
15.
Immun Inflamm Dis ; 10(7): e648, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35759230

RESUMO

INTRODUCTION: The skin immune system is tightly regulated to prevent inappropriate inflammation in response to harmless environmental substances. This regulation is actively maintained by mechanisms including cytokines and cell surface receptors and its loss results in inflammatory disease. In the case of psoriasis, inappropriate immune activation leads to IL-17-driven chronic inflammation, but molecular mechanisms underlying this loss of regulation are not well understood. Immunoglobulin family member CD200 and its receptor, CD200R1, are important regulators of inflammation. Therefore, we determined if this pathway is dysregulated in psoriasis, and how this affects immune cell activity. METHODS: Human skin biopsies were examined by quantitative polymerase chain reaction, flow cytometry, and immunohistochemistry. The role of CD200R1 in regulating psoriasis-like skin inflammation was examined using CD200R1 blocking antibodies in mouse psoriasis models. CD200R1 blocking antibodies were also used in an in vivo neutrophil recruitment assay and in vitro assays to examine macrophage, innate lymphoid cell, γδ T cell, and neutrophil activity. RESULTS: We reveal that CD200 and signaling via CD200R1 are reduced in non-lesional psoriasis skin. In mouse models of psoriasis CD200R1 was shown to limit psoriasis-like inflammation by enhancing acanthosis, CCL20 production and neutrophil recruitment, but surprisingly, macrophage function and IL-17 production were not affected, and neutrophil reactive oxygen species production was reduced. CONCLUSION: Collectively, these data show that CD200R1 affects neutrophil function and limits inflammatory responses in healthy skin by restricting neutrophil recruitment. However, the CD200 pathway is reduced in psoriasis, resulting in a loss of immune control, and increased neutrophil recruitment in mouse models. In conclusion, we highlight CD200R1:CD200 as a pathway that might be targeted to dampen inflammation in patients with psoriasis.


Assuntos
Interleucina-17 , Psoríase , Animais , Anticorpos Bloqueadores , Antígenos CD/metabolismo , Humanos , Imunidade Inata , Inflamação/metabolismo , Linfócitos/metabolismo , Camundongos , Infiltração de Neutrófilos , Receptores de Orexina/genética , Receptores de Orexina/metabolismo
16.
Transl Psychiatry ; 12(1): 122, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35338110

RESUMO

G protein-coupled receptor (GPCR) heterodimers are new targets for the treatment of depression. Increasing evidence supports the importance of serotonergic and orexin-producing neurons in numerous physiological processes, possibly via a crucial interaction between 5-hydroxytryptamine 1A receptor (5-HT1AR) and orexin receptor 1 (OX1R). However, little is known about the function of 5-HT1AR/OX1R heterodimers. It is unclear how the transmembrane domains (TMs) of the dimer affect its function and whether its modulation mediates antidepressant-like effects. Here, we examined the mechanism of 5-HT1AR/OX1R dimerization and downstream G protein-dependent signaling. We found that 5-HT1AR and OX1R form constitutive heterodimers that induce novel G protein-dependent signaling, and that this heterodimerization does not affect recruitment of ß-arrestins to the complex. In addition, we found that the structural interface of the active 5-HT1AR/OX1R dimer transforms from TM4/TM5 in the basal state to TM6 in the active conformation. We also used mutation analyses to identify key residues at the interface (5-HT1AR R1514.40, 5-HT1AR Y1985.41, and OX1R L2305.54). Injection of chronic unpredictable mild stress (CUMS) rats with TM4/TM5 peptides improved their depression-like emotional status and decreased the number of endogenous 5-HT1AR/OX1R heterodimers in the rat brain. These antidepressant effects may be mediated by upregulation of BDNF levels and enhanced phosphorylation and activation of CREB in the hippocampus and medial prefrontal cortex. This study provides evidence that 5-HT1AR/OX1R heterodimers are involved in the pathological process of depression. Peptides including TMs of the 5-HT1AR/OX1R heterodimer interface are candidates for the development of compounds with fast-acting antidepressant-like effects.


Assuntos
Receptor 5-HT1A de Serotonina , Animais , Antidepressivos , Depressão/genética , Depressão/metabolismo , Receptores de Orexina/genética , Fosforilação , Ratos , Receptor 5-HT1A de Serotonina/metabolismo , Transdução de Sinais/genética
17.
Biol Psychiatry ; 91(9): 841-852, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35279280

RESUMO

BACKGROUND: Stress produces differential behavioral responses through select molecular modifications to specific neurocircuitry elements. The orexin (Orx) system targets key components of this neurocircuitry in the basolateral amygdala (BLA). METHODS: We assessed the contribution of intra-BLA Orx1 receptors (Orx1Rs) in the expression of stress-induced phenotypes of mice. Using the Stress Alternatives Model, a social stress paradigm that produces two behavioral phenotypes, we characterized the role of intra-BLA Orx1R using acute pharmacological inhibition (SB-674042) and genetic knockdown (AAV-U6-Orx1R-shRNA) strategies. RESULTS: In the BLA, we observed that Orx1R (Hcrtr1) messenger RNA is predominantly expressed in CamKIIα+ glutamatergic neurons and rarely in GABAergic (gamma-aminobutyric acidergic) cells. While there is a slight overlap in Hcrtr1 and Orx2 receptor (Hcrtr2) messenger RNA expression in the BLA, we find that these receptors are most often expressed in separate cells. Antagonism of intra-BLA Orx1R after phenotype formation shifted behavioral expression from stress-sensitive (Stay) to stress-resilient (Escape) responses, an effect that was mimicked by genetic knockdown. Acute inhibition of Orx1R in the BLA also reduced contextual and cued fear freezing responses in Stay animals. This phenotype-specific behavioral change was accompanied by biased molecular transcription favoring Hcrtr2 over Hcrtr1 and Mapk3 over Plcb1 cell signaling cascades and enhanced Bdnf messenger RNA. CONCLUSIONS: Functional reorganization of intra-BLA gene expression is produced by antagonism of Orx1R, which promotes elevated Hcrtr2, greater Mapk3, and increased Bdnf expression. Together, these results provide evidence for a receptor-driven mechanism that balances pro- and antistress responses within the BLA.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Receptores de Orexina , Animais , Ansiedade/metabolismo , Complexo Nuclear Basolateral da Amígdala/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Camundongos , Receptores de Orexina/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
18.
Nat Methods ; 19(2): 231-241, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35145320

RESUMO

Orexins (also called hypocretins) are hypothalamic neuropeptides that carry out essential functions in the central nervous system; however, little is known about their release and range of action in vivo owing to the limited resolution of current detection technologies. Here we developed a genetically encoded orexin sensor (OxLight1) based on the engineering of circularly permutated green fluorescent protein into the human type-2 orexin receptor. In mice OxLight1 detects optogenetically evoked release of endogenous orexins in vivo with high sensitivity. Photometry recordings of OxLight1 in mice show rapid orexin release associated with spontaneous running behavior, acute stress and sleep-to-wake transitions in different brain areas. Moreover, two-photon imaging of OxLight1 reveals orexin release in layer 2/3 of the mouse somatosensory cortex during emergence from anesthesia. Thus, OxLight1 enables sensitive and direct optical detection of orexin neuropeptides with high spatiotemporal resolution in living animals.


Assuntos
Encéfalo/metabolismo , Imagem Molecular/métodos , Receptores de Orexina/genética , Orexinas/análise , Proteínas Recombinantes/metabolismo , Animais , Comportamento Animal , Feminino , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Receptores de Orexina/metabolismo , Orexinas/genética , Orexinas/farmacologia , Fótons , Proteínas Recombinantes/genética , Reprodutibilidade dos Testes , Sono/fisiologia
19.
J Hum Genet ; 67(6): 377-380, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35034961

RESUMO

Excessive daytime sleepiness is characterized by a persistent feeling of having trouble staying awake, typically with inappropriate sleep episodes. Orexin (hypocretin) is a neuropeptide that regulates sleep-wake cycles and rapid eye movement sleep. Several large-scale genome-wide association studies (GWASs) in European populations have found genetic variants in orexin receptor-1 (OX1R) and -2 (OX2R) that are associated with sleep traits including daytime sleepiness. To identify genetic variants associated with daytime sleepiness, we performed an association study of genetic variants in prepro-orexin, OX1R, and OX2R in 14,329 Japanese individuals from the Tohoku Medical Megabank Project cohort. A genetic variant in OX2R was significantly associated with self-reported daytime sleepiness after Bonferroni correction (rs188018846: P = 8.4E-05). In addition, a missense variant in OX2R identified by the European GWASs showed a nominally significant association with daytime sleepiness in a Japanese population (p.Ile308Val, rs2653349: P = 0.044). Multiple genetic variants in OX2R can affect daytime sleepiness in general populations.


Assuntos
Distúrbios do Sono por Sonolência Excessiva , Estudo de Associação Genômica Ampla , Receptores de Orexina/metabolismo , Distúrbios do Sono por Sonolência Excessiva/epidemiologia , Distúrbios do Sono por Sonolência Excessiva/genética , Humanos , Japão/epidemiologia , Receptores de Orexina/genética , Orexinas/genética , Autorrelato
20.
J Biol Chem ; 298(1): 101521, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34952004

RESUMO

CD200R1 is an inhibitory surface receptor expressed in microglia and blood macrophages. Microglial CD200R1 is known to control neuroinflammation by keeping the microglia in resting state, and therefore, tight regulation of its expression is important. CCAAT/enhancer-binding protein ß (CEBPß) is the known regulator of CD200R1 transcription. In the present study, our specific intention was to find a possible posttranscriptional regulatory mechanism of CD200R1 expression. Here we investigated a novel regulatory mechanism of CD200R1 expression following exposure to an environmental stressor, arsenic, combining in silico analysis, in vitro, and in vivo experiments, as well as validation in human samples. The in silico analysis and in vitro studies with primary neonatal microglia and BV2 microglia revealed that arsenic demethylates the promoter of a microRNA, miR-129-5p, thereby increasing its expression, which subsequently represses CD200R1 by binding to its 3'-untranslated region and shuttling the CD200R1 mRNA to the cytoplasmic-processing body in mouse microglia. The role of miR-129-5p was further validated in BALB/c mouse by stereotaxically injecting anti-miR-129. We found that anti-miR-129 reversed the expression of CD200R1, as well as levels of inflammatory molecules IL-6 and TNF-α. Experiments with a CD200R1 siRNA-induced loss-of-function mouse model confirmed an miR-129-5p→CD200R1→IL-6/TNF-α signaling axis. These main findings were replicated in a human cell line and validated in human samples. Taken together, our study revealed miR-129-5p as a novel posttranscriptional regulator of CD200R1 expression with potential implications in neuroinflammation and related complications.


Assuntos
Arsênio , MicroRNAs , Doenças Neuroinflamatórias , Receptores de Orexina , Regiões 3' não Traduzidas , Animais , Antagomirs/metabolismo , Interleucina-6/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Microglia/metabolismo , Doenças Neuroinflamatórias/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA