Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Tradit Chin Med ; 41(6): 927-934, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34939389

RESUMEN

OBJECTIVE: To investigate the protective effects of Zuogui Wan (ZGW) on bone loss induced by ovariectomy (OVX) and its mechanism via orexin-A and orexin receptors in the osteoporosis rat model. METHODS: Fifty Sprague-Dawley female rats were randomly divided into sham-operated (sham) group and four OVX subgroups. Rats subjected to sham and OVX were treated with the vehicle (OVX, 1 mL/100 g weight, n = 10), 17ß-estradiol (E2, 50 µg*kg-1*d-1), and ZGW at the doses of 2.3 (ZGW-L) and 4.6 (ZGW-H) g/kg/day lyophilized powder daily for 3 months, respectively. The serum biochemical parameters of 17ß-estrogen (17ß-E2), tartrate-resistant acid phosphatase (TRACP-5b) and bone alkaline phosphatase (BALP) were measured by enzyme-linked immunosorbent assay. Hematoxylin-eosin staining was used to detect the changes in the morphological structure in bones. Microcomputed tomography was used to evaluate the bone mineral density and microarchitecture of the distal femur. The gene or protein expression of orexin-A, orexin receptor 1 (OX1R), orexin receptor 2 (OX2R), osteoprotegerin (OPG) and receptor activator of nuclear factor-κB ligand (RANKL) were assayed by either quantitative polymerase chain reaction or Western blot analysis. RESULTS: Compared with the OVX group, ZGW could reduce the serum level of TRACP-5b and increased the serum levels of BALP and17ß-E2 (P < 0.01). Meanwhile, ZGW could prevent bone loss and improved bone trabecular microarchitecture by increasing the trabeculae structure thickness and trabecular number, and arranging the trabeculae structure properly. Compared with the OVX group, it was upregulated for the orexin-A and OX2R mRNA or protein expression from the hypothalamus and tibiae, and OPG in the tibiae of ZGW groups (P < 0.01, < 0.05), while downregulated for the OX1R mRNA and protein expression in the tibiae and hypothalamus and RANKL from the tibiae (P < 0.01). CONCLUSION: ZGW exhibited a protective effect for PMOP that may be mediated via orexin-A and orexin receptors regulation.


Asunto(s)
Hueso Esponjoso , Osteoporosis , Animales , Densidad Ósea , Hueso Esponjoso/metabolismo , Medicamentos Herbarios Chinos , Femenino , Humanos , Receptores de Orexina/genética , Orexinas/genética , Orexinas/farmacología , Orexinas/uso terapéutico , Osteoporosis/etiología , Osteoporosis/genética , Ovariectomía , Ratas , Ratas Sprague-Dawley , Microtomografía por Rayos X
2.
Biomed Khim ; 67(5): 411-417, 2021 Sep.
Artículo en Ruso | MEDLINE | ID: mdl-34730554

RESUMEN

Orexin and its receptors are involved in the mechanisms of pathological craving for alcohol and psychoactive drugs. The orexin system is also involved in the mechanisms of non-chemical forms of addiction: binge eating and gambling. The aim of this work was to study the level of orexin receptor mRNA in the hypothalamus, hippocampus, and prefrontal cortex of rats prone to impulsivity in behavior in a model for studying the elements of gambling addiction (a variant of the Iowa Gambling Task test). Brain structures were isolated on the 22nd day of the experiment. The expression of the OX1R gene was higher in the hypothalamus by 122% and in the hippocampus by 149% in rats that preferred to receive a high reward, but with a low probability as compared with a group of animals that preferred a low level of reinforcement, but with a 100% probability. In the prefrontal cortex, on the contrary, no significant changes were observed in the level of OX1R mRNA. The level of OX2R mRNA insignificantly changed in the hypothalamus, hippocampus, and prefrontal cortex of rats prone to impulsivity in behavior. The data indicate involvement of OX1R in the hypothalamus and hippocampus in mechanisms mediating impulsive behavior and the choice of the significance of positive reinforcement in terms of its varying strength and probability.


Asunto(s)
Encéfalo , Hipotálamo , Animales , Conducta Impulsiva , Receptores de Orexina/genética , ARN Mensajero/genética , Ratas
3.
Artículo en Inglés | MEDLINE | ID: mdl-32296386

RESUMEN

Orexin receptors (OXRs) play a critical regulatory role in central control of food intake, maintenance of sleeping states, energy metabolism, and neuroendocrine homeostasis. However, most previous studies have focused on the sleep-promoting functions of OXRs in human beings, while their potential value in enhancing food intake for livestock breeding has not been fully exploited. In this study, we successfully cloned porcine orexin 2 receptor (pOX2R) complementary DNA and constructed four pOX2R mutants (P10S, P11T, V308I, and T401I) by site-directed mutagenesis, and their functional expressions were further confirmed through Western blotting analysis. Pharmacological characteristics of pOX2R and their mutants were further investigated. These results showed that the P10S, P11T, and T401I mutants had decreased cAMP signaling with orexin A, whereas only the P11T mutant decreased under the stimulation of orexin B. Besides, only P10S displayed a decreased calcium release in response to both orexin ligands. Importantly, these mutants exhibited decreased phosphorylation levels of ERK1/2, p38, and CREB to some degree compared with wild-type pOX2R. Collectively, these findings highlight the critical role of these mutations in pOX2R signaling and expand our understanding of molecular and pharmacological characterization of pOX2R.


Asunto(s)
Receptores de Orexina/metabolismo , Orexinas/farmacología , Porcinos , Animales , Clonación Molecular , Células HEK293 , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación , Receptores de Orexina/química , Receptores de Orexina/genética , Orexinas/metabolismo , Filogenia , Conformación Proteica , Transducción de Señal/efectos de los fármacos , Porcinos/genética , Porcinos/metabolismo
4.
Behav Brain Res ; 384: 112455, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32044404

RESUMEN

Understanding the mechanisms underlying memory is essential for the treatment of post-traumatic stress disorder (PTSD). Orexin, as a lateral hypothalamic (LH) neuropeptide, interferes with the stages of memory, primarily through the orexin receptor1 (Orx1R). The aim of this study was to evaluate the effects of amygdala Orx1R in the acquisition and extinction processes of PTSD modeled in animals. In three experiments, rats were divided into three groups: control (Naïve), shock (receiving a foot shock), and PTSD (experiencing Single prolonged stress (SPS) method). The first experiment aimed to evaluate LH activity in PTSD modeled rats. The second and third experiments aimed to evaluate the effects of Orx1R in the acquisition and extinction of fear memory in PTSD modeled animals. SB334867 (SB) or its solvent was microinjected into the amygdala and the rats were subjected to conditioning thereafter. In the second group, we used a single injection after conditioning. In the third group, we used three consecutive injections (one after each memory test). Some behaviors and Orx1R expression were evaluated. The freezing response was significantly longer in the PTSD group than on the control. Similarly, anxiety and sensitized fear were also intensified. CFos expression levels in LH was higher in the PTSD group. Inhibition of Orx1R in the amygdala significantly decreased memory acquisition, diminished anxiety, and decreased the sensitized fear in the SB group. Applying SB to the amygdala after each fear memory test significantly decreased freezing. Expression of Orx1R was significantly higher following fear conditioning. These results indicate a likely involvement of the orexin and amygdalar Orx1R in memory acquisition and in extinction of PTSD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Extinción Psicológica/fisiología , Memoria/fisiología , Receptores de Orexina/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Animales , Ansiedad/genética , Ansiedad/metabolismo , Conducta Animal , Benzoxazoles/farmacología , Modelos Animales de Enfermedad , Prueba de Laberinto Elevado , Miedo , Reacción Cataléptica de Congelación , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Naftiridinas/farmacología , Prueba de Campo Abierto , Antagonistas de los Receptores de Orexina/farmacología , Receptores de Orexina/efectos de los fármacos , Receptores de Orexina/genética , Orexinas/metabolismo , ARN Mensajero , Ratas , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/fisiopatología , Urea/análogos & derivados , Urea/farmacología
5.
Biomed Res Int ; 2019: 2389485, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31346513

RESUMEN

Acute alcohol exposure induces unconscious condition such as coma whose main physical manifestation is the loss of righting reflex (LORR). Xingnaojing Injection (XNJI), which came from Chinese classic formula An Gong Niu Huang Pill, is widely used for consciousness disorders in China, such as coma. Although XNJI efficiently shortened the duration of LORR induced by acute ethanol, it remains unknown how XNJI acts on ethanol-induced coma (EIC). We performed experiments to examine the effects of XNJI on orexin and adenosine (AD) signaling in the lateral hypothalamic area (LHA) in EIC rats. Results showed that XNJI reduced the duration of LORR, which implied that XNJI promotes recovery form coma. Microdialysis data indicated that acute ethanol significantly increased AD release in the LHA but had no effect on orexin A levels. The qPCR results displayed a significant reduction in the Orexin-1 receptors (OX1R) expression with a concomitant increase in the A1 receptor (A1R) and equilibrative nucleoside transporter type 1 (ENT1) expression in EIC rats. In contrast, XNJI reduced the extracellular AD levels but orexin A levels remained unaffected. XNJI also counteracted the downregulation of the OX1R expression and upregulation of A1R and ENT1 expression caused by EIC. As for ADK expression, XNJI but not ethanol, displayed an upregulation in the LHA in EIC rats. Based on these results, we suggest that XNJI promotes arousal by inhibiting adenosine neurotransmission via reducing AD level and the expression of A1R and ENT1.


Asunto(s)
Proteínas Portadoras/genética , Coma/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Receptor de Adenosina A1/genética , Adenosina/genética , Adenosina/metabolismo , Animales , Coma/inducido químicamente , Coma/genética , Coma/patología , Tranportador Equilibrativo 1 de Nucleósido , Etanol/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Receptores de Orexina/genética , Orexinas/genética , Orexinas/metabolismo , Ratas , Reflejo de Enderezamiento/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Vigilia/efectos de los fármacos
6.
Keio J Med ; 68(1): 27, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30905886

RESUMEN

Although sleep is a ubiquitous behavior in animal species with well-developed central nervous systems, many aspects in the neurobiology of sleep remain mysterious. Our discovery of orexin, a hypothalamic neuropeptide involved in the maintenance of wakefulness, has triggered an intensive research examining the exact role of the orexinergic and other neural pathways in the regulation of sleep/wakefulness. The orexin receptor antagonist suvorexant, which specifically block the endogenous waking system, has been approved as a new drug to treat insomnia. Also, since the sleep disorder narcolepsy-cataplexy is caused by orexin deficiency, orexin receptor agonists are expected to provide mechanistic therapy for narcolepsy; they will likely be also useful for treating excessive sleepiness due to other etiologies.Despite the fact that the executive neurocircuitry and neurochemistry for sleep/wake switching has been increasingly revealed in recent years, the mechanism for homeostatic regulation of sleep, as well as the neural substrate for "sleepiness" (sleep need), remains unknown. To crack open this black box, we have initiated a large-scale forward genetic screen of sleep/wake phenotype in mice based on true somnographic (EEG/EMG) measurements. We have so far screened >8,000 heterozygous ENU-mutagenized founders and established a number of pedigrees exhibiting heritable and specific sleep/wake abnormalities. By combining linkage analysis and the next-generation whole exome sequencing, we have molecularly identified and verified the causal mutation in several of these pedigrees. Biochemical and neurophysiological analyses of these mutations are underway. Since these dominant mutations cause strong phenotypic traits, we expect that the mutated genes will provide new insights into the elusive pathway regulating sleep/wakefulness. Indeed, through a systematic cross-comparison of the Sleepy mutants and sleep-deprived mice, we have recently found that the cumulative phosphorylation state of a specific set of mostly synaptic proteins may be the molecular substrate of sleep need.


Asunto(s)
Cataplejía/genética , Narcolepsia/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Sueño/fisiología , Vigilia/fisiología , Animales , Azepinas/farmacología , Cataplejía/tratamiento farmacológico , Cataplejía/fisiopatología , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Hipotálamo/fisiopatología , Ratones , Ratones Transgénicos , Mutación , Narcolepsia/tratamiento farmacológico , Narcolepsia/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Antagonistas de los Receptores de Orexina/farmacología , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Polisomnografía , Fármacos Inductores del Sueño/farmacología , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Triazoles/farmacología
7.
Mol Neurobiol ; 56(8): 5315-5331, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30603957

RESUMEN

Inhibitors of DNA methylation and orexin type-1 receptor antagonists modulate the neurobiological effects driving drugs of abuse and natural reinforcers by activating common brain structures of the mesolimbic reward system. In this study, we applied a self-administration paradigm to assess the involvement of factors regulating DNA methylation processes and satiety or appetite signals. These factors include Dnmts and Tets, miR-212/132, orexins, and orx-R1 genes. The study focused on dopamine projection areas such as the prefrontal cortex (PFCx) and caudate putamen (CPu) and in the hypothalamus (HP) that is interconnected with the reward system. Striking changes were observed in response to both reinforcers, but differed depending on contingent and non-contingent delivery. Expression also differed in the PFCx and the CPu. Cocaine and food induced opposite effects on Dnmt3a expression in both brain structures, whereas they repressed both miRs to a different extent, without affecting their primary transcript in the CPu. Unexpectedly, orexin mRNAs were found in the CPu, suggesting a transport from their transcription site in the HP. The orexin receptor1 gene was found to be induced by cocaine in the PFCx, consistent with a regulation by DNA methylation. Global levels of 5-methylcytosines in the PFCx were not significantly altered by cocaine, suggesting that it is rather their distribution that contributes to long-lasting behaviors. Together, our data demonstrate that DNA methylation regulating factors are differentially altered by cocaine and food. At the molecular level, they support the idea that neural circuits activated by both reinforcers do not completely overlap.


Asunto(s)
Encéfalo/metabolismo , Cocaína/administración & dosificación , Metilación de ADN/genética , Alimentos , Orexinas/metabolismo , Autoadministración , Animales , Condicionamiento Operante , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Conducta Alimentaria , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Péptidos/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Putamen/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , ADN Metiltransferasa 3B
8.
Proc Natl Acad Sci U S A ; 115(45): E10740-E10747, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348769

RESUMEN

Orexin (also known as hypocretin) neurons in the hypothalamus play an essential role in sleep-wake control, feeding, reward, and energy homeostasis. The likelihood of anesthesia and sleep sharing common pathways notwithstanding, it is important to understand the processes underlying emergence from anesthesia. In this study, we investigated the role of the orexin system in anesthesia emergence, by specifically activating orexin neurons utilizing the designer receptors exclusively activated by designer drugs (DREADD) chemogenetic approach. With injection of adeno-associated virus into the orexin-Cre transgenic mouse brain, we expressed the DREADD receptor hM3Dq specifically in orexin neurons and applied the hM3Dq ligand clozapine to activate orexin neurons. We monitored orexin neuronal activities by c-Fos staining and whole-cell patch-clamp recording and examined the consequence of orexin neuronal activation via EEG recording. Our results revealed that the orexin-DREADD mice with activated orexin neurons emerged from anesthesia with significantly shorter latency than the control mice. As an indication of reduced pain sensitivity, these orexin-DREADD mice took longer to respond to the 55 °C thermal stimuli in the hot plate test and exhibited significantly less frequent licking of the formalin-injected paw in the formalin test. Our study suggests that approaches to activate the orexin system can be beneficial in postoperative recovery.


Asunto(s)
Periodo de Recuperación de la Anestesia , Hipotálamo/metabolismo , Neuronas/metabolismo , Receptores de Orexina/genética , Orexinas/genética , Dolor/genética , Anestésicos por Inhalación , Animales , Clozapina/farmacología , Dependovirus/genética , Dependovirus/metabolismo , Electroencefalografía , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Calor , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiopatología , Isoflurano , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/patología , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Dolor/fisiopatología , Dolor/prevención & control , Dimensión del Dolor , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Antagonistas de la Serotonina/farmacología , Técnicas Estereotáxicas
9.
Acupunct Med ; 36(5): 312-318, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29669795

RESUMEN

OBJECTIVES: Decreased lung function in chronic obstructive pulmonary disease (COPD) is correlated with abnormal excitability of the respiratory centre where orexin neuropeptides from the hypothalamus are responsible for regulating respiration. We hypothesised that improvements in pulmonary function with electroacupuncture (EA) may be related to orexins in a rat model of COPD. METHODS: The COPD model was established by cigarette smoke exposure and lipopolysaccharide injection. Modelled rats received EA at BL13 and ST36 for two weeks, after which lung function was tested. Orexin levels in the hypothalamus and medulla were detected by ELISA, while mRNA/protein expression and localisation of orexins and their receptors were investigated using real time PCR, Western blotting and immunohistochemistry, respectively. RESULTS: The decrease in lung function observed in COPD rats was improved after EA treatment. Orexin levels in the hypothalamus and medulla were significantly higher in COPD rats than in normal rats, but were significantly reduced in the EA-treated group. There was a negative correlation between orexin content and lung function. In the hypothalamus, mRNA and protein expression and immunoreactivity of orexins were significantly higher in the COPD group than in the normal group, but a significant decrease was observed after EA. In the medulla, the expression and immunoreactivity of orexin receptors were significantly higher in the COPD group than in the normal group, but a significant decrease was observed after EA. CONCLUSIONS: The positive effect of EA on pulmonary function in COPD rats may be related to downregulation of orexins and their receptors in the medulla.


Asunto(s)
Electroacupuntura , Hipotálamo/metabolismo , Bulbo Raquídeo/metabolismo , Orexinas/genética , Enfermedad Pulmonar Obstructiva Crónica/terapia , Puntos de Acupuntura , Animales , Modelos Animales de Enfermedad , Humanos , Pulmón/metabolismo , Masculino , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Auton Neurosci ; 210: 34-43, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29246398

RESUMEN

In peripheral tissues, aldosterone alters expression of multiple genes, including the clock gene Period 1 (Per1), 11 beta-hydroxysteroid dehydrogenase-2 (11-HSD2), and α-ENAC, the epithelial sodium channel subunit. We evaluated the impact of chronic aldosterone exposure (DOCA) and salt intake on nocturnal changes in gene expression in the male Sprague Dawley rat brain. Additionally, genes associated with the orexin (ORX) system were also evaluated based on the role of this neuropeptide in arousal, feeding and hypertension and an interconnection with Per1 expression. DOCA/salt treatment increased saline intake primarily at night, elevated arterial pressure and lowered heart rate. In the medulla oblongata, DOCA/salt upregulated Per1, 11-HSD2, and α-ENAC expression independent of time of day, but did not change ORX receptor type 1 (ORX-R1) or type 2 (ORX-R2) expression. ORX-R1, and ORX-R2 expression in the medulla did however correlate with Per1 expression following DOCA/salt treatment but not in controls. In the hypothalamus, DOCA/salt treatment upregulated Per1, ORX-A, and ORX-R2 expression, in general, and Per1 and ORX-A expression at night. ORX-A, ORX-R1 and ORX-R2 expression in the hypothalamus correlated with Per1 expression following DOCA/salt but not in controls. These findings demonstrate for the first time that DOCA/salt hypertension modulates ORX gene expression in the brain and suggest that changes in expression in the ORX system may occur directly or indirectly via aldosterone-induced changes in Per1 expression. Our findings also build on emerging evidence that monitoring gene expression during both the day and night is critical to understanding the role of specific genes in hypertension.


Asunto(s)
Desoxicorticosterona/farmacología , Hipertensión/patología , Hipotálamo/efectos de los fármacos , Bulbo Raquídeo/efectos de los fármacos , Orexinas/metabolismo , Proteínas Circadianas Period/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Ritmo Circadiano/fisiología , Modelos Animales de Enfermedad , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/inducido químicamente , Hipotálamo/metabolismo , Masculino , Bulbo Raquídeo/metabolismo , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Proteínas Circadianas Period/genética , Ratas , Ratas Sprague-Dawley , Cloruro de Sodio/farmacología
11.
Proc Natl Acad Sci U S A ; 114(22): 5731-5736, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28507129

RESUMEN

Narcolepsy-cataplexy is a debilitating disorder of sleep/wakefulness caused by a loss of orexin-producing neurons in the lateroposterior hypothalamus. Genetic or pharmacologic orexin replacement ameliorates symptoms in mouse models of narcolepsy-cataplexy. We have recently discovered a potent, nonpeptide OX2R-selective agonist, YNT-185. This study validates the pharmacological activity of this compound in OX2R-transfected cells and in OX2R-expressing neurons in brain slice preparations. Intraperitoneal, and intracerebroventricular, administration of YNT-185 suppressed cataplexy-like episodes in orexin knockout and orexin neuron-ablated mice, but not in orexin receptor-deficient mice. Peripherally administered YNT-185 also promotes wakefulness without affecting body temperature in wild-type mice. Further, there was no immediate rebound sleep after YNT-185 administration in active phase in wild-type and orexin-deficient mice. No desensitization was observed after repeated administration of YNT-185 with respect to the suppression of cataplexy-like episodes. These results provide a proof-of-concept for a mechanistic therapy of narcolepsy-cataplexy by OX2R agonists.


Asunto(s)
Compuestos de Anilina/farmacología , Benzamidas/farmacología , Cataplejía/tratamiento farmacológico , Narcolepsia/tratamiento farmacológico , Receptores de Orexina/agonistas , Orexinas/metabolismo , Trastornos del Sueño del Ritmo Circadiano/tratamiento farmacológico , Promotores de la Vigilia/uso terapéutico , Vigilia/efectos de los fármacos , Compuestos de Anilina/química , Animales , Benzamidas/química , Modelos Animales de Enfermedad , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Orexina/genética , Orexinas/genética , Técnicas de Placa-Clamp , Sueño/efectos de los fármacos
12.
EBioMedicine ; 18: 311-319, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28377228

RESUMEN

BACKGROUND: Neurophysiological and behavioral processes regulated by hypocretin (orexin) are severely affected in depression. However, alterations in hypocretin have so far not been studied in the human brain. We explored the hypocretin system changes in the hypothalamus and cortex in depression from male and female subjects. METHODS: We quantified the differences between depression patients and well-matched controls, in terms of hypothalamic hypocretin-1 immunoreactivity (ir) and hypocretin receptors (Hcrtr-receptors)-mRNA in the anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex. In addition, we determined the alterations in the hypocretin system in a frequently used model for depression, the chronic unpredictable mild stress (CUMS) rat. RESULTS: i) Compared to control subjects, the amount of hypocretin-immunoreactivity (ir) was significantly increased in female but not in male depression patients; ii) hypothalamic hypocretin-ir showed a clear diurnal fluctuation, which was absent in depression; iii) male depressive patients who had committed suicide showed significantly increased ACC Hcrt-receptor-2-mRNA expression compared to male controls; and iv) female but not male CUMS rats showed a highly significant positive correlation between the mRNA levels of corticotropin-releasing hormone and prepro-hypocretin in the hypothalamus, and a significantly increased Hcrt-receptor-1-mRNA expression in the frontal cortex compared to female control rats. CONCLUSIONS: The clear sex-related change found in the hypothalamic hypocretin-1-ir in depression should be taken into account in the development of hypocretin-targeted therapeutic strategies.


Asunto(s)
Trastorno Depresivo Mayor/patología , Orexinas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Trastorno Bipolar/metabolismo , Trastorno Bipolar/patología , Corticosterona/sangre , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Trastorno Depresivo Mayor/metabolismo , Modelos Animales de Enfermedad , Femenino , Giro del Cíngulo/metabolismo , Humanos , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Corteza Prefrontal/metabolismo , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales
13.
Am J Physiol Heart Circ Physiol ; 312(4): H808-H817, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28159808

RESUMEN

Orexin neurons, and activation of orexin receptors, are generally thought to be sympathoexcitatory; however, the functional connectivity between orexin neurons and a likely sympathetic target, the hypothalamic spinally projecting neurons (SPNs) in the paraventricular nucleus of the hypothalamus (PVN) has not been established. To test the hypothesis that orexin neurons project directly to SPNs in the PVN, channelrhodopsin-2 (ChR2) was selectively expressed in orexin neurons to enable photoactivation of ChR2-expressing fibers while examining evoked postsynaptic currents in SPNs in rat hypothalamic slices. Selective photoactivation of orexin fibers elicited short-latency postsynaptic currents in all SPNs tested (n = 34). These light-triggered responses were heterogeneous, with a majority being excitatory glutamatergic responses (59%) and a minority of inhibitory GABAergic (35%) and mixed glutamatergic and GABAergic currents (6%). Both glutamatergic and GABAergic responses were present in the presence of tetrodotoxin and 4-aminopyridine, suggesting a monosynaptic connection between orexin neurons and SPNs. In addition to generating postsynaptic responses, photostimulation facilitated action potential firing in SPNs (current clamp configuration). Glutamatergic, but not GABAergic, postsynaptic currents were diminished by application of the orexin receptor antagonist almorexant, indicating orexin release facilitates glutamatergic neurotransmission in this pathway. This work identifies a neuronal circuit by which orexin neurons likely exert sympathoexcitatory control of cardiovascular function.NEW & NOTEWORTHY This is the first study to establish, using innovative optogenetic approaches in a transgenic rat model, that there are robust heterogeneous projections from orexin neurons to paraventricular spinally projecting neurons, including excitatory glutamatergic and inhibitory GABAergic neurotransmission. Endogenous orexin release modulates glutamatergic, but not GABAergic, neurotransmission in these pathways.


Asunto(s)
Hipotálamo/citología , Hipotálamo/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Acetamidas/farmacología , Potenciales de Acción/fisiología , Animales , Animales Modificados Genéticamente , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ácido Glutámico/metabolismo , Hipotálamo/diagnóstico por imagen , Isoquinolinas/farmacología , Masculino , Vías Nerviosas/citología , Vías Nerviosas/diagnóstico por imagen , Optogenética , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Núcleo Hipotalámico Paraventricular/diagnóstico por imagen , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Médula Espinal/diagnóstico por imagen , Ácido gamma-Aminobutírico/metabolismo
14.
Gen Comp Endocrinol ; 242: 66-73, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26631456

RESUMEN

The peptides orexin A (OxA) and orexin B (OxB) deriving from a common precursor molecule, prepro-orexin, by proteolytic cleavage, bind the two G-coupled OX1 and OX2 receptors. While OX1 selectively binds OxA, OX2 shows similar affinity for both orexins. Firstly discovered in the hypothalamus, orexins and their receptors have been found in other brain regions as well as in peripheral tissues of mammals, thus resulting involved in the regulation of a broad variety of physiological functions. While the functional localization of OxA and OX1 in the mammalian genital tract has been already described, the expression of OxB and OX2 and their potential role in the reproductive functions remain to be explored. Here, we investigated the presence of OxB and OX2 in the rat testis by immunohistochemical and biochemical analyses. The results definitely demonstrated the localization of OxB and OX2 in pachytene and second spermatocytes as well as in spermatids at all stages of the cycle of the seminiferous epithelium. The expression of both OX2 mRNA and protein in the rat testis was also established by RT-PCR and Western blotting, respectively. The analysis of the molecular mechanism of action of OxB in the rat testis showed that OxB, in contrast with OxA, is unable to promote steroidogenesis. These results translate into the regulation of diverse biological actions by OxA and OxB in the male gonad.


Asunto(s)
Receptores de Orexina/metabolismo , Orexinas/metabolismo , Testículo/metabolismo , Animales , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neuropéptidos/metabolismo , Receptores de Orexina/genética , Orexinas/genética , Ratas , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/metabolismo , Espermátides/metabolismo
15.
Peptides ; 88: 55-61, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27988352

RESUMEN

Orexins, also known as hypocretins, play a regulatory role in the sleep-wake cycle by activating orexin receptors. Previous animal studies have shown that sleep deprivation can elevate orexinergic peptide levels. However, the relationship between insomnia disorder and orexin-A levels in humans has not been explored. In the current study, we examined plasma orexin-A levels in patients with insomnia disorder and in normal sleepers. We also studied the possible mechanisms underlying changes in orexin-A levels between the study groups; this included investigations of prepro-orexin and orexin receptor gene polymorphisms as well as exploration of other variables. We measured plasma orexin-A levels in 228 patients with insomnia disorder and 282 normal sleepers. The results indicated that the patients with insomnia disorder had significantly higher orexin-A levels than normal sleepers (63.42±37.56 vs. 54.84±23.95pg/ml). A positive relationship was detected between orexin-A level and age in patients with insomnia disorder. Orexin-A levels were elevated in relation to course of insomnia, as well as in relation to increased Insomnia Severity Index score. None of the evaluated prepro-orexin gene single nucleotide polymorphisms were informative between the two study populations. After sequencing all orexin receptor exons, one variation (rs2271933) in the OX1R gene and one variation (rs2653349) in the OX2R gene were found. However, no significant differences were found in either genotypic or allelic frequency distributions between the two study groups. It is suggested that the increased plasma orexin-A levels in patients with insomnia disorder are associated with the course and severity of insomnia, but not with prepro-orexin and orexin receptor gene polymorphisms.


Asunto(s)
Receptores de Orexina/genética , Orexinas/sangre , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Adulto , Anciano , Animales , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Hipotálamo/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Orexina/sangre , Polimorfismo de Nucleótido Simple , Sueño/genética , Sueño/fisiología , Privación de Sueño/genética , Privación de Sueño/fisiopatología , Trastornos del Inicio y del Mantenimiento del Sueño/sangre , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología
16.
Eur J Neurosci ; 44(10): 2795-2806, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27562056

RESUMEN

Shiftworkers run an increased risk of developing metabolic disorders, presumably as a result of disturbed circadian physiology. Eating at a time-of-day that is normally dedicated to resting and fasting, may contribute to this association. The hypothalamus is the key brain area that integrates different inputs, including environmental time information from the central biological clock in the suprachiasmatic nuclei, with peripheral information on energy status to maintain energy homeostasis. The orexin system within the lateral hypothalamus is an important output of the suprachiasmatic nuclei involved in the control of sleep/wake behavior and glucose homeostasis, among other functions. In this study, we tested the hypothesis that feeding during the rest period disturbs the orexin system as a possible underlying contributor to metabolic health problems. Male Wistar rats were exposed to an 8-week protocol in which food was available ad libitum for 24-h, for 12-h during the light phase (i.e., unnatural feeding time) or for 12-h during the dark phase (i.e., restricted feeding, but at the natural time-of-day). Animals forced to eat at an unnatural time, i.e., during the light period, showed no changes in orexin and orexin-receptor gene expression in the hypothalamus, but the rhythmic expression of clock genes in the lateral hypothalamus was absent in these animals. Light fed animals did show adverse changes in whole-body physiology and internal desynchronization of muscle and liver clock and metabolic gene expression. Eating at the 'wrong' time-of-day thus causes internal desynchronization at different levels, which in the long run may disrupt body physiology.


Asunto(s)
Ciclos de Actividad , Ritmo Circadiano , Conducta Alimentaria , Hígado/fisiología , Músculo Esquelético/fisiología , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Ratas , Ratas Wistar
17.
Nat Commun ; 7: 11395, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-27102565

RESUMEN

The lateral hypothalamus (LH) controls energy balance. LH melanin-concentrating-hormone (MCH) and orexin/hypocretin (OH) neurons mediate energy accumulation and expenditure, respectively. MCH cells promote memory and appropriate stimulus-reward associations; their inactivation disrupts energy-optimal behaviour and causes weight loss. However, MCH cell dynamics during wakefulness are unknown, leaving it unclear if they differentially participate in brain activity during sensory processing. By fiberoptic recordings from molecularly defined populations of LH neurons in awake freely moving mice, we show that MCH neurons generate conditional population bursts. This MCH cell activity correlates with novelty exploration, is inhibited by stress and is inversely predicted by OH cell activity. Furthermore, we obtain brain-wide maps of monosynaptic inputs to MCH and OH cells, and demonstrate optogenetically that VGAT neurons in the amygdala and bed nucleus of stria terminalis inhibit MCH cells. These data reveal cell-type-specific LH dynamics during sensory integration, and identify direct neural controllers of MCH neurons.


Asunto(s)
Redes Reguladoras de Genes , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Receptores de Orexina/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/metabolismo , Animales , Mapeo Encefálico , Metabolismo Energético/genética , Conducta Exploratoria/fisiología , Tecnología de Fibra Óptica , Regulación de la Expresión Génica , Hormonas Hipotalámicas/genética , Hipotálamo/citología , Masculino , Melaninas/genética , Ratones , Ratones Transgénicos , Neuronas/clasificación , Neuronas/citología , Optogenética , Receptores de Orexina/genética , Orexinas/genética , Técnicas de Placa-Clamp , Hormonas Hipofisarias/genética , Núcleos Septales/citología , Núcleos Septales/metabolismo , Técnicas Estereotáxicas , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores , Vigilia/genética
18.
Iran J Allergy Asthma Immunol ; 15(1): 20-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26996108

RESUMEN

Orexin A and B are hypothalamic peptides with a wide variety of effects such as anti-inflammation and neuroprotection. Impaired function of orexin system has been reported in some neurodegenerative diseases like Parkinson, Huntington and Alzheimer. In this study, the mRNA expression levels of some hypothalamic peptides were investigated in C57BL/6 female mice with experimental autoimmune encephalomyelitis (EAE). Animals were randomly divided into two control and EAE groups. EAE was induced by administration of myelin oligodendrocyte glycoprotein (MOG) with complete Ferund's adjuvant and pertussis toxin. Twenty-first days following immunization, mice were decapitated to remove the brains. Then, the expression profiles of prepro-orexin, orexin 1 receptors (OX1R) and orexin 2 receptors (OX2R) in hypothalamic region were assessed using real-time PCR method. In this study, we found a considerable increase in the mRNA expression of OX1R and OX2R following EAE induction in C57BL/6 mice. Elevation levels of OX1R and OX2R following EAE induction suggest that alteration in orexinergic system may involve in pathogenesis of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Receptores de Orexina/genética , ARN Mensajero/genética , Animales , Conducta Animal , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/psicología , Femenino , Adyuvante de Freund , Hipotálamo/metabolismo , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Receptores de Orexina/metabolismo , Toxina del Pertussis , ARN Mensajero/metabolismo , Regulación hacia Arriba
19.
Gen Comp Endocrinol ; 239: 62-70, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-26562300

RESUMEN

Orexin A (OXA), a hypothalamic neuropeptide, and its receptor (OX1R) are primarily expressed in lateral hypothalamus and are involved in the control of various biological functions. Expressions of OXA and OX1R have also been reported in peripheral organs like gastrointestinal and genital tracts. In the present study, expressions of OXA and OX1R have been observed in the testis of Parkes strain neonatal mice by semi-quantitative RT-PCR and western blot analyses. Immunohistochemical study also revealed their presence on spermatogonia, Sertoli cells and in the interstitium of the testis. In order to understand the role of OXA and OX1R in testicular development, an in vitro study was also performed. For this, binding of OXA to OX1R was blocked using OX1R specific antagonist, SB-334867. Eighteen mice were sacrificed and their testes were cultured in complete media containing vehicle and two doses (0.1 and 4.0µg/ml media) of SB-334867 for 72h in CO2 incubator at 37°C. At the end of culture period, testes were used for western blot and RT-PCR analyses to study the expression of various markers of gonadal development, such as steroidogenic factor 1 (SF-1), Wilms' tumor 1 (Wt1), Mullerian inhibiting substance (MIS) and stem cell factor (SCF). Further, expressions of OXA, OX1R and glucose transporter 3 (GLUT 3) were also studied. A marked increase in the expression of SF-1 and a decrease in the expression of Wt1 at both transcript and protein levels were noted, while there was a decrease in the expression of SCF and MIS at transcript level at both doses of the antagonist; this suggests that blockage of OXA binding to OX1R by SB-334867 affects testicular development. The decrease in expressions of OXA, OX1R and GLUT 3 in the test is in response to both doses of the antagonist points to their down-regulation causing inefficient uptake of glucose by the testicular cells, thereby affecting gonadal development. In conclusion, our results suggest that the binding of OXA to OX1R is important for the development of the testis.


Asunto(s)
Receptores de Orexina/genética , Receptores de Orexina/fisiología , Orexinas/genética , Orexinas/fisiología , Testículo/metabolismo , Animales , Animales Recién Nacidos , Hormona Antimülleriana/farmacología , Benzoxazoles/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Naftiridinas , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Testículo/efectos de los fármacos , Distribución Tisular , Urea/análogos & derivados , Urea/farmacología
20.
Scand J Gastroenterol ; 51(2): 157-67, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26199984

RESUMEN

OBJECTIVES: Gastric electrical stimulation (GES) has great potential for the treatment of obesity. We investigated the impact of chronic GES on the alteration of adipose tissue and the regulation of neuropeptide Y (NPY), orexin (OX), α-melanocyte-stimulating hormone (α-MSH) and oxytocin (OXT), and their receptors in several tissues. MATERIAL AND METHODS: Most of the experiments included three groups of diet-induced obesity rats: (1) sham-GES (SGES); (2) GL-6mA (GES with 6 mA, 4 ms, 40 Hz, 2 s on, 3 s off at lesser curvature); and (3) SGES-PF (SGES rats receiving pair feeding to match the consumption of GL-6mA rats). Chronic GES was applied for 2 h every day for 4 weeks. During treatment with GES, food intake and body weight were monitored weekly. The alteration of epididymal fat weight, gastric emptying, and expression of peptides and their receptors in several tissues were determined. RESULTS: GL-6mA was more potent than SGES-PF in decreasing body weight gain, epididymal fat tissue weight, adipocyte size and gastric emptying. Chronic GES significantly altered NPY, OX, α-MSH and OXT and their receptors in the hypothalamus, adipose tissue and stomach. CONCLUSIONS: Chronic GES effectively leads to weight loss by reducing food intake, fat tissue weight and gastric emptying. NPY, α-MSH, orexin and OXT, and their receptors in the hypothalamus, adipose tissue and stomach appear to be involved in the anti-obesity effects of chronic GES.


Asunto(s)
Terapia por Estimulación Eléctrica/métodos , Mucosa Gástrica/metabolismo , Hipotálamo/metabolismo , Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Obesidad/terapia , ARN Mensajero/metabolismo , Adipocitos/patología , Animales , Modelos Animales de Enfermedad , Ingestión de Alimentos , Electrodos Implantados , Epidídimo , Vaciamiento Gástrico , Ghrelina/metabolismo , Grasa Intraabdominal/patología , Leptina/genética , Masculino , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Receptores de Orexina/genética , Orexinas/metabolismo , Oxitocina/genética , Oxitocina/metabolismo , Proopiomelanocortina/genética , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 3 , Receptores Acoplados a Proteínas G/genética , Receptores de Melanocortina/genética , Receptores de Neuropéptido/genética , Receptores de Neuropéptido Y/genética , Receptores de Oxitocina/genética , Pérdida de Peso , alfa-MSH/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA