Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.690
Filtrar
1.
Bioorg Med Chem ; 112: 117892, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39236468

RESUMEN

Dual orexin receptor antagonists (DORAs) are approved for the treatment of sleep onset and/or sleep maintenance insomnia. In the present disclosure, we report the discovery of a new class of DORAs designed to treat sleep disorders requiring a fast onset and a short duration of action (<4 h). We used early human pharmacokinetic-pharmacodynamic (PK-PD) predictions and in vivo experiments to identify DORAs eliciting this specific hypnotic profile. A high-throughput screening campaign revealed hits based on a rarely precedented tricyclic pyrazolidine scaffold. After unsuccessful structure-activity-relationship (SAR) studies on this hit series, a scaffold hopping exercise, aimed at reducing the molecular complexity of the tricyclic scaffold, resulted in the discovery of the 2-acyl-1-biarylmethylpyrazolidine series. SAR studies on this achiral series gave rise to the lead compound DORA 42. In vitro and in vivo parameters of DORA 42, and its PK-PD simulation for human use are detailed.


Asunto(s)
Descubrimiento de Drogas , Antagonistas de los Receptores de Orexina , Pirazoles , Relación Estructura-Actividad , Humanos , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/química , Antagonistas de los Receptores de Orexina/síntesis química , Pirazoles/química , Pirazoles/farmacología , Pirazoles/síntesis química , Animales , Estructura Molecular , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/síntesis química , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/farmacocinética , Receptores de Orexina/metabolismo , Ratas , Relación Dosis-Respuesta a Droga , Masculino
2.
Cephalalgia ; 44(9): 3331024241281493, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39233656

RESUMEN

BACKGROUND: We wished to explore possible sexual dimorphism in mechanisms sensitizing or activating meningeal nociceptors that can promote the headache phase of migraine. METHODS: Male and female C57BL6J mice received either supradural orexin B and an inflammatory mediator cocktail (IM) with migraine-like pain behaviors and photophobia recorded. Expression of orexin 2 receptor (OX2R) in trigeminal ganglion (TG) and phosphorylated extracellular signal-regulated kinases (ERK) levels in trigeminal nucleus caudalis (TNC) were evaluated. Orexin B-induced excitability of TG cells was assessed with patch-clamp electrophysiology. Intranasal delivery of CRISPR/Cas9 plasmids was used to edit the expression of OX2R in the TG. RESULTS: Supradural orexin B induced migraine-like pain behaviors, photophobia and increased TNC ERK phosphorylation exclusively in males. Blockade of orexin signaling with supradural suvorexant, a dual orexin receptor antagonist, prevented, but did not reverse, migraine-like pain in males induced by supradural IM cocktail. OX2R expression was higher in male TG and orexin B increased TG neuron excitability in males. Intranasal OX2R CRISPR/Cas9 reduced TG receptor expression and orexin B-induced TNC ERK phosphorylation and prevented migraine-like pain induced by supradural orexin B in males. CONCLUSIONS: Our studies reveal a male-specific mechanism of TG nociceptor sensitization and migraine-like pain behavior mediated by orexin B/OX2R signaling. Sexually dimorphic mechanisms of trigeminal nociceptor sensitization and activation offer opportunities to improve patient outcomes by considering patient sex and may influence clinical trial design and interpretation.


Asunto(s)
Ratones Endogámicos C57BL , Trastornos Migrañosos , Receptores de Orexina , Ganglio del Trigémino , Animales , Masculino , Femenino , Ratones , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/fisiopatología , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/efectos de los fármacos , Meninges/efectos de los fármacos , Meninges/metabolismo , Caracteres Sexuales , Orexinas/metabolismo
3.
J Neuroinflammation ; 21(1): 229, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39294682

RESUMEN

BACKGROUND: Overactivated microglia are a key contributor to Parkinson's disease (PD) by inducing neuroinflammation. CD200R1, a membrane glycoprotein mainly found on microglia, is crucial for maintaining quiescence with its dysregulation linked to microglia's abnormal activation. We and other groups have reported a decline in CD200R1 levels in several neurological disorders including PD. However, the mechanism regulating CD200R1 expression and the specific reasons for its reduction in PD remain largely unexplored. Given the pivotal role of transcription factors in gene expression, this study aimed to elucidate the transcriptional regulation of CD200R1 and its implications in PD. METHODS: The CD200R1 promoter core region was identified via luciferase assays. Potential transcription factors were predicted using the UCSC ChIP-seq database and JASPAR. NFKB1 binding to the CD200R1 core promoter was substantiated through electrophoretic mobility shift and chromatin immunoprecipitation assays. Knocking-down or overexpressing NFKB1 validated its regulatory effect on CD200R1. Correlation between decreased CD200R1 and deficient NFKB1 was studied using Genotype-Tissue Expression database. The clinical samples of the peripheral blood mononuclear cells were acquired from 44 PD patients (mean age 64.13 ± 9.78, 43.2% male, median Hoehn-Yahr stage 1.77) and 45 controls (mean age 64.70 ± 9.41, 52.1% male). NFKB1 knockout mice were utilized to study the impact of NFKB1 on CD200R1 expression and to assess their roles in PD pathophysiology. RESULTS: The study identified the CD200R1 core promoter region, located 482 to 146 bp upstream of its translation initiation site, was directly regulated by NFKB1. Significant correlation between NFKB1 and CD200R1 expression was observed in human PMBCs. Both NFKB1 and CD200R1 were significantly decreased in PD patient samples. Furthermore, NFKB1-/- mice exhibited exacerbated microglia activation and dopaminergic neuron loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. CONCLUSION: Our study identified that NFKB1 served as a direct regulator of CD200R1. Reduced NFKB1 played a critical role in CD200R1 dysregulation and subsequent microglia overactivation in PD. These findings provide evidence that targeting the NFKB1-CD200R1 axis would be a novel therapeutic strategy for PD.


Asunto(s)
Subunidad p50 de NF-kappa B , Receptores de Orexina , Enfermedad de Parkinson , Animales , Humanos , Ratones , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Masculino , Femenino , Persona de Mediana Edad , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p50 de NF-kappa B/genética , Anciano , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Ratones Endogámicos C57BL , Regulación de la Expresión Génica , Microglía/metabolismo , Regiones Promotoras Genéticas
4.
Sci Rep ; 14(1): 20838, 2024 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242684

RESUMEN

Narcolepsy type 1 (NT1) is associated with severe loss of orexin neurons and characterized by symptoms including excessive daytime sleepiness and cataplexy. Current medications indicated for NT1 often show limited efficacy, not addressing the full spectrum of symptoms, demonstrating a need for novel drugs. We discovered a parenteral orexin receptor 2 (OX2R) agonist, danavorexton, and an orally available OX2R agonist, TAK-994; both improving NT1 phenotypes in mouse models and individuals with NT1. However, danavorexton has limited oral availability and TAK-994 has a risk of off-target liver toxicity. To avoid off-target-based adverse events, a highly potent molecule with low effective dose is preferred. Here, we show that a novel OX2R-selective agonist, TAK-861 [N-{(2S,3R)-4,4-Difluoro-1-(2-hydroxy-2-methylpropanoyl)-2-[(2,3',5'-trifluoro[1,1'-biphenyl]-3-yl)methyl]pyrrolidin-3-yl}ethanesulfonamide], activates OX2R with a half-maximal effective concentration of 2.5 nM and promotes wakefulness at 1 mg/kg in mice and monkeys, suggesting ~ tenfold higher potency and lower effective dosage than TAK-994. Similar to TAK-994, TAK-861 substantially ameliorates wakefulness fragmentation and cataplexy-like episodes in orexin/ataxin-3 and orexin-tTA;TetO DTA mice (NT1 mouse models). Compared with modafinil, TAK-861 induces highly correlated brain-wide neuronal activation in orexin-tTA;TetO DTA mice, suggesting efficient wake-promoting effects. Thus, TAK-861 has potential as an effective treatment for individuals with hypersomnia disorders including narcolepsy, potentially with a favorable safety profile.


Asunto(s)
Modelos Animales de Enfermedad , Narcolepsia , Receptores de Orexina , Vigilia , Animales , Narcolepsia/tratamiento farmacológico , Receptores de Orexina/agonistas , Receptores de Orexina/metabolismo , Vigilia/efectos de los fármacos , Ratones , Administración Oral , Fenotipo , Masculino , Humanos
5.
Peptides ; 180: 171280, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39159833

RESUMEN

The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.


Asunto(s)
Nivel de Alerta , Neuropéptidos , Receptores de Orexina , Orexinas , Recompensa , Orexinas/metabolismo , Humanos , Animales , Nivel de Alerta/fisiología , Neuropéptidos/metabolismo , Receptores de Orexina/metabolismo , Antagonistas de los Receptores de Orexina/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Motivación/fisiología , Neuronas/metabolismo
6.
Neuropeptides ; 107: 102463, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39180799

RESUMEN

Studies have indicated that stress-related symptoms can lead to hormonal and neural changes, affecting the pain threshold and nociceptive behaviors. The precise role of orexin receptors (OX1r and OX2r) in stress-induced analgesia (SIA) remains an inquiry yet to be comprehensively elucidated. The current investigation aimed to assess the impact of acute immobilization restraint stress on pain-related behavioral responses after administering antagonists targeting OX1r and OX2r in a rat model using the tail-flick test. After a period of five to seven days post-stereotaxic surgery in CA1, the baseline tail-flick latency (TFL) was recorded for each animal. Subsequently, rats were unilaterally administered varying doses of the OX1r antagonist (SB334867; 1, 3, 10, and 30 nmol), the OX2r antagonist (TCS OX2 29; 1, 3, 10, and 30 nmol), or a vehicle (0.5 µl solution containing 12% DMSO) through an implanted cannula. Following a 5-min interval, the animals were subjected to a restraint stress (RS) lasting for 3 h. The tail-flick test was conducted after the stress exposure, and the TFLs were assessed at 60-min intervals. The findings of this study revealed that RS elicits antinociceptive responses in the tail-flick test. Microinjection of OX1r and OX2r antagonists into the CA1 attenuated RS-induced analgesia during the tail-flick test. Furthermore, the results underscored the preeminent role of OX2 receptors in modulating SIA. In conclusion, the orexin system localized within the hippocampal CA1 region may, in part, contribute to the manifestation of SIA in the context of acute pain.


Asunto(s)
Benzoxazoles , Región CA1 Hipocampal , Naftiridinas , Antagonistas de los Receptores de Orexina , Receptores de Orexina , Restricción Física , Estrés Psicológico , Animales , Receptores de Orexina/metabolismo , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/administración & dosificación , Masculino , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Estrés Psicológico/metabolismo , Ratas , Benzoxazoles/farmacología , Benzoxazoles/administración & dosificación , Naftiridinas/farmacología , Urea/análogos & derivados , Urea/farmacología , Urea/administración & dosificación , Isoquinolinas/farmacología , Isoquinolinas/administración & dosificación , Ratas Sprague-Dawley , Analgésicos/farmacología , Analgésicos/administración & dosificación , Piridinas/farmacología , Piridinas/administración & dosificación , Dolor/tratamiento farmacológico , Dolor/metabolismo , Aminopiridinas , Sulfonamidas
7.
Bioorg Med Chem ; 110: 117823, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38964170

RESUMEN

Molecular imaging using positron emission tomography (PET) can serve as a promising tool for visualizing biological targets in the brain. Insights into the expression pattern and the in vivo imaging of the G protein-coupled orexin receptors OX1R and OX2R will further our understanding of the orexin system and its role in various physiological and pathophysiological processes. Guided by crystal structures of our lead compound JH112 and the approved hypnotic drug suvorexant bound to OX1R and OX2R, respectively, we herein describe the design and synthesis of two novel radioligands, [18F]KD23 and [18F]KD10. Key to the success of our structural modifications was a bioisosteric replacement of the triazole moiety with a fluorophenyl group. The 19F-substituted analog KD23 showed high affinity for the OX1R and selectivity over OX2R, while the high affinity ligand KD10 displayed similar Ki values for both subtypes. Radiolabeling starting from the respective pinacol ester precursors resulted in excellent radiochemical yields of 93% and 88% for [18F]KD23 and [18F]KD10, respectively, within 20 min. The new compounds will be useful in PET studies aimed at subtype-selective imaging of orexin receptors in brain tissue.


Asunto(s)
Receptores de Orexina , Tomografía de Emisión de Positrones , Receptores de Orexina/metabolismo , Ligandos , Humanos , Relación Estructura-Actividad , Estructura Molecular , Radiofármacos/química , Radiofármacos/síntesis química , Descubrimiento de Drogas , Triazoles/química , Triazoles/síntesis química , Triazoles/farmacología , Radioisótopos de Flúor/química , Antagonistas de los Receptores de Orexina/química , Antagonistas de los Receptores de Orexina/síntesis química , Antagonistas de los Receptores de Orexina/farmacología
8.
Sci Rep ; 14(1): 15964, 2024 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987562

RESUMEN

Pathological proteins including tau are produced in neurons and released into interstitial fluid (ISF) in a neural activity-dependent manner during wakefulness. Pathological proteins in ISF can be removed from the brain via the glymphatic pathway during nighttime. Thus, in individuals with Alzheimer's disease (AD) that have dysregulated sleep/wake rhythm, application of orexin receptor 2 (OX2R) agonists during daytime could recover the efflux of pathological proteins to ISF and indirectly promote the glymphatic pathway by improving the quality of nighttime sleep after proper daytime arousal, resulting in increased removal of these proteins from the brain. We investigated this hypothesis using OX-201, a novel OX2R-selective agonist with a 50% effective concentration of 8.0 nM. Diurnal rhythm of tau release into hippocampal ISF correlated well with neuronal activity and wakefulness in wild-type mice. In both wild-type and human P301S tau transgenic mice, OX-201 induced wakefulness and promoted tau release into hippocampal ISF. Human P301S tau transgenic mice, tested under our conditions, showed longer wakefulness time, which differs from individuals with AD. OX-201 treatment over 2 months did not alter hippocampal tau levels. Although further studies are required, at a minimum OX2R agonists may not exacerbate tau accumulation in individuals with tauopathy, including AD.


Asunto(s)
Enfermedad de Alzheimer , Hipocampo , Ratones Transgénicos , Receptores de Orexina , Proteínas tau , Animales , Proteínas tau/metabolismo , Ratones , Receptores de Orexina/metabolismo , Receptores de Orexina/agonistas , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Vigilia/efectos de los fármacos , Masculino , Líquido Extracelular/metabolismo , Líquido Extracelular/efectos de los fármacos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos
9.
Cell Mol Life Sci ; 81(1): 288, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38970689

RESUMEN

Orexinergic neurons are critically involved in regulating arousal, wakefulness, and appetite. Their dysfunction has been associated with sleeping disorders, and non-peptide drugs are currently being developed to treat insomnia and narcolepsy. Yet, no light-regulated agents are available to reversibly control their activity. To meet this need, a photoswitchable peptide analogue of the endogenous neuroexcitatory peptide orexin-B was designed, synthesized, and tested in vitro and in vivo. This compound - photorexin - is the first photo-reversible ligand reported for orexin receptors. It allows dynamic control of activity in vitro (including almost the same efficacy as orexin-B, high nanomolar potency, and subtype selectivity to human OX2 receptors) and in vivo in zebrafish larvae by direct application in water. Photorexin induces dose- and light-dependent changes in locomotion and a reduction in the successive induction reflex that is associated with sleep behavior. Molecular dynamics calculations indicate that trans and cis photorexin adopt similar bent conformations and that the only discriminant between their structures and activities is the positioning of the N-terminus. This, in the case of the more active trans isomer, points towards the OX2 N-terminus and extra-cellular loop 2, a region of the receptor known to be involved in ligand binding and recognition consistent with a "message-address" system. Thus, our approach could be extended to several important families of endogenous peptides, such as endothelins, nociceptin, and dynorphins among others, that bind to their cognate receptors through a similar mechanism: a "message" domain involved in receptor activation and signal transduction, and an "address" sequence for receptor occupation and improved binding affinity.


Asunto(s)
Luz , Receptores de Orexina , Orexinas , Pez Cebra , Receptores de Orexina/metabolismo , Receptores de Orexina/química , Animales , Orexinas/metabolismo , Humanos , Locomoción/efectos de los fármacos , Simulación de Dinámica Molecular , Larva/metabolismo , Larva/efectos de los fármacos , Células HEK293 , Ligandos
10.
Sleep Med ; 121: 303-314, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39047304

RESUMEN

Sleep is considered closely related to cognitive function, and cognitive impairment is the main clinical manifestation of Alzheimer's disease (AD). Sleep disturbance in AD patients is more severe than that in healthy elderly individuals. Additionally, sleep deprivation reportedly increases the activity of the hypothalamic orexin system and the risk of AD. To investigate whether intervention with the orexin system can improve sleep disturbance in AD and its impact on AD pathology. In this study, six-month-old amyloid precursor protein/presenilin 1 mice were subjected to six weeks of chronic sleep deprivation and injected intraperitoneally with almorexant, a dual orexin receptor antagonist (DORA), to investigate the effects and mechanisms of sleep deprivation and almorexant intervention on learning and memory in mice with AD. We found that sleep deprivation aggravated learning and memory impairment and increased brain ß-amyloid (Aß) deposition in mice with AD. The application of almorexant can increase the total sleep time of sleep-deprived mice and reduce cognitive impairment and Aß deposition, which is related to the improvement in Aquaporin-4 polarity. Thus, DORA may be an effective strategy for delaying the progression of AD patients by improving the sleep disturbances.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Trastornos de la Memoria , Ratones Transgénicos , Antagonistas de los Receptores de Orexina , Privación de Sueño , Animales , Privación de Sueño/complicaciones , Privación de Sueño/tratamiento farmacológico , Ratones , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Precursor de Proteína beta-Amiloide/genética , Acetamidas/farmacología , Acetamidas/uso terapéutico , Masculino , Péptidos beta-Amiloides/metabolismo , Aprendizaje/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Receptores de Orexina/metabolismo , Presenilina-1/genética
11.
Food Funct ; 15(17): 8661-8673, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39056112

RESUMEN

Background: A high-fat diet (HFD) is generally associated with an increased risk of mental disorders that constitute a sizeable worldwide health. A HFD results in the gut microbiota-brain axis being altered and linked to mental disorders. Hypocretin-1, which can promote appetite, has been previously confirmed to be associated with depression. However, no exact relationship has been found for hypocretin between depression and HFDs. Methods: Adult male SD rats were randomly assigned to either a HFD or a normal diet for eight weeks, followed by behavioral tests and plasma biochemical analyses. Then, we investigated the protein and mRNA levels of inflammation-related factors in the hippocampus. We also observed morphological changes in brain microglia and lipid accumulation. Additionally, metagenomic and metabolomic analyses of gut microbiomes were performed. 3T3-L1 cells were utilized in vitro to investigate the impact of hypocretin receptor 1 antagonists (SB334867) on lipid accumulation. To consider the connection between the brain and adipose tissue, we used a conditioned medium (CM) treated with 3T3-L1 cells to observe the activation and phagocytosis of BV2 cells. Following a 12-week period of feeding a HFD to C57BL/6 mice, a three-week intervention period was initiated during which the administration of SB334867 was observed. This was followed by a series of assessments, including monitoring of body weight changes and emotional problems, as well as attention to plasma biochemical levels and microglial cell phenotypes in the brain. Results: The HFD rats displayed anxiety and depressive-like behaviors. HFD rats exhibited increased plasma HDL, LDL, and TC levels. A HFD also causes an increase in hypocretin-1 and hypocretin-2 in the hypothalamus. Metagenomics and metabolomics revealed that the HFD caused an increase in the relative abundance of associated inflammatory bacteria and decreased the abundance of anti-inflammatory and bile acid metabolites. Compared with the CTR group, hippocampal microglia in the HFD group were significantly activated and accompanied by lipid deposition. At the same time, protein and mRNA expression levels of inflammation-related factors were increased. We found that SB334867 could significantly reduce lipid accumulation in 3T3-L1 cells after differentiation. The expression of inflammatory factors decreased in the SB334867 group. The administration of SB334867 was found to reverse the adverse effects of the HFD on body weight, depressive-like behaviour and anxiety-like mood. Furthermore, this treatment was associated with improvements in plasma biochemical levels and a reduction in the number of microglia in the brain. Conclusions: In summary, our results demonstrated that a HFD induced anxiety and depressive-like behaviors, which may be linked to the increased hypocretin-1 level and lipid accumulation. Supplementation with SB334867 improved the above. These observations highlight the possibility of hypocretin-1 inducing the risk of HFD-associated emotional dysfunctions.


Asunto(s)
Depresión , Dieta Alta en Grasa , Microbioma Gastrointestinal , Inflamación , Ratones Endogámicos C57BL , Receptores de Orexina , Orexinas , Ratas Sprague-Dawley , Animales , Masculino , Ratones , Ratas , Células 3T3-L1 , Benzoxazoles , Depresión/metabolismo , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Inflamación/metabolismo , Microglía/metabolismo , Microglía/efectos de los fármacos , Naftiridinas , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Orexinas/metabolismo , Fenotipo , Urea/análogos & derivados
12.
Behav Brain Res ; 472: 115133, 2024 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-38960330

RESUMEN

The complicated relevance between stress and pain has been identified. Neurotransmitters and neuropeptides of various brain areas play a role in this communication. Pain inhibitory response is known as stress-induced analgesia (SIA). The studies demonstrated that the nucleus accumbens (NAc) is critical in modulating pain. As a neuropeptide, orexin is crucially involved in initiating behavioral and physiological responses to threatening and unfeeling stimuli. However, the role of the orexin receptors of the NAc area after exposure to restraint stress (RS) as acute physical stress in the modulation of acute pain is unclear. One hundered twenty adult male albino Wistar rats (230-250 g) were used. Animals were unilaterally implanted with cannulae above the NAc. The SB334867 and TCS OX2 29 were used as antagonists for OX1r and OX2r, respectively. Different doses of the antagonists (1, 3, 10, and 30 nmol/0.5 µl DMSO) were microinjected intra-NAc five minutes before exposure to RS (3 hours). Then, the tail-flick test as a model of acute pain was performed, and the nociceptive threshold (Tail-flick latency; TFL) was measured in 60-minute time set intervals. According to this study's findings, the antinociceptive effects of RS in the tail-flick test were blocked during intra-NAc administration of SB334867 or TCS OX2 29. The RS as acute stress increased TFL and deceased pain-like behavior responses. The 50 % effective dose values of the OX1r and OX2r antagonists were 12.82 and 21.64 nmol, respectively. The result demonstrated contribution of the OX1r into the NAc was more remarkable than that of the OX2r on antinociceptive responses induced by the RS. Besides, in the absence of RS, the TFL was attenuated. The current study's data indicated that OX1r and OX2r into the NAc induced pain modulation responses during RS in acute pain. In conclusion, the findings revealed the involvement of intra-NAc orexin receptors in improving SIA.


Asunto(s)
Dolor Agudo , Benzoxazoles , Naftiridinas , Núcleo Accumbens , Antagonistas de los Receptores de Orexina , Receptores de Orexina , Ratas Wistar , Restricción Física , Estrés Psicológico , Urea , Animales , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Masculino , Receptores de Orexina/metabolismo , Benzoxazoles/farmacología , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/administración & dosificación , Urea/análogos & derivados , Urea/farmacología , Urea/administración & dosificación , Dolor Agudo/fisiopatología , Dolor Agudo/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Naftiridinas/farmacología , Isoquinolinas/farmacología , Isoquinolinas/administración & dosificación , Ratas , Piridinas/farmacología , Piridinas/administración & dosificación , Orexinas/farmacología , Orexinas/metabolismo , Relación Dosis-Respuesta a Droga , Dimensión del Dolor/efectos de los fármacos , Aminopiridinas , Sulfonamidas
13.
PLoS One ; 19(6): e0306099, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38917189

RESUMEN

Orexin-mediated stimulation of orexin receptors 1/2 (OX[1/2]R) may stimulate the diaphragm and genioglossus muscle via activation of inspiratory neurons in the pre-Bötzinger complex, which are critical for the generation of inspiratory rhythm, and phrenic and hypoglossal motoneurons. Herein, we assessed the effects of OX2R-selective agonists TAK-925 (danavorexton) and OX-201 on respiratory function. In in vitro electrophysiologic analyses using rat medullary slices, danavorexton and OX-201 showed tendency and significant effect, respectively, in increasing the frequency of inspiratory synaptic currents of inspiratory neurons in the pre-Bötzinger complex. In rat medullary slices, both danavorexton and OX-201 significantly increased the frequency of inspiratory synaptic currents of hypoglossal motoneurons. Danavorexton and OX-201 also showed significant effect and tendency, respectively, in increasing the frequency of burst activity recorded from the cervical (C3-C5) ventral root, which contains axons of phrenic motoneurons, in in vitro electrophysiologic analyses from rat isolated brainstem-spinal cord preparations. Electromyogram recordings revealed that intravenous administration of OX-201 increased burst frequency of the diaphragm and burst amplitude of the genioglossus muscle in isoflurane- and urethane-anesthetized rats, respectively. In whole-body plethysmography analyses, oral administration of OX-201 increased respiratory activity in free-moving mice. Overall, these results suggest that OX2R-selective agonists enhance respiratory function via activation of the diaphragm and genioglossus muscle through stimulation of inspiratory neurons in the pre-Bötzinger complex, and phrenic and hypoglossal motoneurons. OX2R-selective agonists could be promising drugs for various conditions with respiratory dysfunction.


Asunto(s)
Diafragma , Nervio Hipogloso , Neuronas Motoras , Receptores de Orexina , Nervio Frénico , Animales , Diafragma/efectos de los fármacos , Diafragma/inervación , Diafragma/fisiología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Receptores de Orexina/agonistas , Receptores de Orexina/metabolismo , Ratas , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiología , Ratones , Masculino , Nervio Hipogloso/efectos de los fármacos , Nervio Hipogloso/fisiología , Ratas Sprague-Dawley , Inhalación , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Isoquinolinas , Piridinas
14.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38892431

RESUMEN

Orexin-A is a neuropeptide product of the lateral hypothalamus that acts on two receptors, OX1R and OX2R. The orexinergic system is involved in feeding, sleep, and pressure regulation. Recently, orexin-A levels have been found to be negatively correlated with renal function. Here, we analyzed orexin-A levels as well as the incidence of SNPs in the hypocretin neuropeptide precursor (HCRT) and its receptors, HCRTR1 and HCRTR2, in 64 patients affected by autosomal dominant polycystic kidney disease (ADPKD) bearing truncating mutations in the PKD1 or PKD2 genes. Twenty-four healthy volunteers constituted the control group. Serum orexin-A was assessed by ELISA, while the SNPs were investigated through Sanger sequencing. Correlations with the main clinical features of PKD patients were assessed. PKD patients showed impaired renal function (mean eGFR 67.8 ± 34.53) and a statistically higher systolic blood pressure compared with the control group (p < 0.001). Additionally, orexin-A levels in PKD patients were statistically higher than those in healthy controls (477.07 ± 69.42 pg/mL vs. 321.49 ± 78.01 pg/mL; p < 0.001). Furthermore, orexin-A inversely correlated with blood pressure (p = 0.0085), while a direct correlation with eGFR in PKD patients was found. None of the analyzed SNPs showed any association with orexin-A levels in PKD. In conclusion, our data highlights the emerging role of orexin-A in renal physiology and its potential relevance to PKD. Further research is essential to elucidate the intricate mechanisms underlying orexin-A signaling in renal function and its therapeutic implications for PKD and associated cardiovascular complications.


Asunto(s)
Receptores de Orexina , Orexinas , Polimorfismo de Nucleótido Simple , Humanos , Orexinas/metabolismo , Orexinas/genética , Masculino , Femenino , Persona de Mediana Edad , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Adulto , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/sangre , Estudios de Casos y Controles , Anciano , Presión Sanguínea , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/sangre
15.
Pharmacol Rev ; 76(5): 625-688, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38902035

RESUMEN

The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.


Asunto(s)
Antagonistas de los Receptores de Orexina , Receptores de Orexina , Humanos , Receptores de Orexina/metabolismo , Receptores de Orexina/fisiología , Animales , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/uso terapéutico , Terminología como Asunto
16.
Aging (Albany NY) ; 16(11): 9558-9568, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38829778

RESUMEN

Osteoarthritis (OA) is one of the most important causes of global disability, and dysfunction of chondrocytes is an important risk factor. The treatment of OA is still a challenge. Orexin-A is a hypothalamic peptide, and its effects in OA are unknown. In this study, we found that exposure to interleukin-1ß (IL-1ß) reduced the expression of orexin-2R, the receptor of orexin-A in TC-28a2 chondrocytes. Importantly, the senescence-associated ß-galactosidase (SA-ß-gal) staining assay demonstrated that orexin-A treatment ameliorates IL-1ß-induced cellular senescence. Importantly, the presence of IL-1ß significantly reduced the telomerase activity of TC-28a2 chondrocytes, which was rescued by orexin-A. We also found that orexin-A prevented IL-1ß-induced increase in the levels of Acetyl-p53 and the expression of p21. It is shown that orexin-A mitigates IL-1ß-induced reduction of sirtuin 3 (SIRT3). Silencing of SIRT3 abolished the protective effects of orexin-A against IL-1ß-induced cellular senescence. These results imply that orexin-A might serve as a promising therapeutic agent for OA.


Asunto(s)
Senescencia Celular , Condrocitos , Interleucina-1beta , Orexinas , Senescencia Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Orexinas/farmacología , Orexinas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Osteoartritis/metabolismo , Osteoartritis/tratamiento farmacológico , Humanos , Sirtuina 3/metabolismo , Sirtuina 3/genética , Animales , Proteína p53 Supresora de Tumor/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Línea Celular
17.
Mol Cell Endocrinol ; 592: 112312, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38866320

RESUMEN

Orexins (OXs) are neuropeptides which regulate various physiological processes. OXs exist in two different forms, mainly orexin A (OXA) and orexin B (OXB) and their effects are mediated via OX1R and OX2R. Presence of OXB and OX2R in mouse testis is also reported. However, the role of OXB/OX2R in the male gonad remains unexplored. Herein we investigated the role of OXB/OX2R system in testicular physiology under in vivo and ex vivo conditions. Adult mice were given a single dose of bilateral intratesticular injection of siRNA targeting OX2R and were sacrificed 96 h post-injection. OX2R-knockdown potentiated serum and intratesticular testosterone levels with up-regulation in the expressions of major steroidogenic proteins. Germ cell proliferation also increased in siRNA-treated mice. Results of the ex vivo experiment also supported the findings of the in vivo study. In conclusion, OX2R may regulate testosterone production and thereby control the fine-tuning between steroidogenesis and germ cell dynamics.


Asunto(s)
Proliferación Celular , Receptores de Orexina , Testículo , Testosterona , Animales , Masculino , Testosterona/metabolismo , Testosterona/sangre , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Testículo/metabolismo , Ratones , Técnicas de Silenciamiento del Gen , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética , Orexinas/metabolismo , Orexinas/genética , Células Intersticiales del Testículo/metabolismo , Células Germinativas/metabolismo
18.
J Psychopharmacol ; 38(7): 647-660, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38888086

RESUMEN

BACKGROUND: The orexin (OX) system has received increasing interest as a potential target for treating substance use disorder. OX transmission in the posterior paraventricular nucleus of the thalamus (pPVT), an area activated by highly salient stimuli that are both reinforcing and aversive, mediates cue- and stress-induced reinstatement of reward-seeking behavior. Oral administration of suvorexant (SUV), a dual OX receptor (OXR) antagonist (DORA), selectively reduced conditioned reinstatement of oxycodone-seeking behavior and stress-induced reinstatement of alcohol-seeking behavior in dependent rats. AIMS: This study tested whether OXR blockade in the pPVT with SUV reduces oxycodone or sweetened condensed milk (SCM) seeking elicited by conditioned cues or stress. METHODS: Male Wistar rats were trained to self-administer oxycodone (0.15 mg/kg, i.v., 8 h/day) or SCM (0.1 ml, 2:1 dilution [v/v], 30 min/day). After extinction, we tested the ability of intra-pPVT SUV (15 µg/0.5 µl) to prevent reinstatement of oxycodone or SCM seeking elicited by conditioned cues or footshock stress. RESULTS: The rats acquired oxycodone and SCM self-administration, and oxycodone intake correlated with signs of physical opioid withdrawal, confirming dependence. Following extinction, the presentation of conditioned cues or footshock elicited reinstatement of oxycodone- and SCM-seeking behavior. Intra-pPVT SUV blocked stress-induced reinstatement of oxycodone seeking but not conditioned reinstatement of oxycodone or SCM seeking or stress-induced reinstatement of SCM seeking. CONCLUSIONS: The results indicate that OXR signaling in the pPVT is critical for stress-induced reinstatement of oxycodone seeking, further corroborating OXRs as treatment targets for opioid use disorder.


Asunto(s)
Comportamiento de Búsqueda de Drogas , Antagonistas de los Receptores de Orexina , Orexinas , Oxicodona , Ratas Wistar , Autoadministración , Estrés Psicológico , Animales , Oxicodona/farmacología , Oxicodona/administración & dosificación , Masculino , Ratas , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Estrés Psicológico/metabolismo , Orexinas/metabolismo , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/administración & dosificación , Señales (Psicología) , Transducción de Señal/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Receptores de Orexina/metabolismo , Receptores de Orexina/efectos de los fármacos , Recompensa , Trastornos Relacionados con Opioides/metabolismo , Núcleos Talámicos de la Línea Media/efectos de los fármacos , Núcleos Talámicos de la Línea Media/metabolismo
19.
Tissue Cell ; 88: 102381, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692160

RESUMEN

Diabetic retinopathy (DR) is established as the primary cause of visual impairment and preventable blindness, posing significant social and economic burdens on healthcare systems worldwide. Oxidative stress has been identified as a major contributor to DR, yet the precise role of the transmembrane glycoprotein CD200R in this context remains elusive. We studied human retinal pigment epithelia ARPE-19 cells to investigate the role of CD200R in high-glucose (HG) induced oxidative stress. Under HG conditions, we found a significant increase in CD200R expression in a time-dependent pattern. Conversely, knockdown of CD200R effectively alleviated oxidative stress and restored cell viability in HG-treated ARPE-19 cells, a phenomenon corroborated by the addition of a reactive oxygen species (ROS) scavenger. Exploration of the AKT/mTOR signaling pathway confirmed its mediating role regarding CD200R knockdown suppression of the expression of key proteins induced by HG conditions. Additionally, we found that the inhibition of mTOR signaling with Rapamycin effectively countered HG-induced oxidative stress in ARPE-19 cells, suggesting a promising therapeutic target against oxidative stress in the context of DR. This study establishes the crucial role of CD200R in HG-induced oxidative stress and identifies potential therapeutic avenues for the treatment of DR.


Asunto(s)
Glucosa , Estrés Oxidativo , Epitelio Pigmentado de la Retina , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Estrés Oxidativo/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Glucosa/farmacología , Línea Celular , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Especies Reactivas de Oxígeno/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología
20.
Zhen Ci Yan Jiu ; 49(5): 441-447, 2024 May 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38764114

RESUMEN

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.


Asunto(s)
Puntos de Acupuntura , Encéfalo , Electroacupuntura , Receptores de Orexina , Dolor , Animales , Humanos , Masculino , Ratones , Encéfalo/metabolismo , Inflamación/terapia , Inflamación/metabolismo , Inflamación/genética , Ratones Endogámicos C57BL , Receptores de Orexina/metabolismo , Receptores de Orexina/genética , Dolor/metabolismo , Dolor/genética , Manejo del Dolor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA