Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 31(1): 458-480, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38015391

RESUMEN

Nonmedical use of modafinil (MOD) led to increased rates of overdose toxicity, road accidents, addiction, withdrawal, suicide, and mental illnesses. The current study aims to determine the probable MOD brain toxicity and elucidate the possible role of selenium (Se) in ameliorating the neurotoxicity in rat models. Fifty-four male Albino rats were randomly assigned into nine groups. The groups were G1 (control negative), G2 (Se0.1), G3 (Se0.2), G4 (MOD300), G5 (MOD600), G6 (Se0.1 + MOD300), G7 (Se0.2 + MOD300), G8 (Se0.1 + MOD600), and G9 (Se0.2 + MOD600). After finishing the experiment, blood and brain tissue were harvested for biochemical and histological investigation. Neurobehavior parameters were assessed. Tissue neurotransmitter levels and oxidative stress markers were assessed. Gene expression of PI3K/Akt/mTOR-GSK3B, orexin, and orexin receptor2 was measured by qRT-PCR. Histological and immunohistochemistry assessments, as well as molecular docking, were carried out. MOD-induced neurobehavioral toxicity exhibited by behavioral and cognitive function impairments, which are associated with decreased antioxidant activities, increased MDA levels, and decreases in neurotransmitter levels. Brain levels of mRNA expression of PI3K, Akt, and mTOR were decreased, while GS3K, orexin, and orexin receptors were significantly elevated. These disturbances were confirmed by histopathological brain changes with increased silver and Bax immunostaining and decreased crystal violet levels. MOD induced neurotoxic effects in a dose-dependent manner. Compared with the MOD groups, SE coadministration significantly attenuates MOD-induced toxic changes. Docking study shows the protective role of Se as an apoptosis inhibitor and inflammation inhibitor. In conclusion, Se could be used as a biologically effective antioxidant compound to protect from MOD neurobehavioral toxicity in Wistar rats by reversing behavioral alterations, inflammation, apoptosis, and oxidative injury.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Selenio , Humanos , Ratas , Masculino , Animales , Selenio/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antioxidantes/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Modafinilo/farmacología , Orexinas/metabolismo , Orexinas/farmacología , Simulación del Acoplamiento Molecular , Ratas Wistar , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Estrés Oxidativo , Inflamación , Apoptosis , Neurotransmisores
2.
Br J Pharmacol ; 181(9): 1474-1493, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38129941

RESUMEN

BACKGROUND AND PURPOSE: We evaluated the hypothesis that central orexin application could counteract motion sickness responses through regulating neural activity in target brain areas. EXPERIMENTAL APPROACH: Thec effects of intracerebroventricular (i.c.v.) injection of orexin-A and SB-334867 (OX1 antagonist) on motion sickness-induced anorexia, nausea-like behaviour (conditioned gaping), hypoactivity and hypothermia were investigated in rats subjected to Ferris wheel-like rotation. Orexin-A responsive brain areas were identified using Fos immunolabelling and were verified via motion sickness responses after intranucleus injection of orexin-A, SB-334867 and TCS-OX2-29 (OX2 antagonist). The efficacy of intranasal application of orexin-A versus scopolamine on motion sickness symptoms in cats was also investigated. KEY RESULTS: Orexin-A (i.c.v.) dose-dependently attenuated motion sickness-related behavioural responses and hypothermia. Fos expression was inhibited in the ventral part of the dorsomedial hypothalamus (DMV) and the paraventricular nucleus (PVN), but was enhanced in the ventral part of the premammillary nucleus ventral part (PMV) by orexin-A (20 µg) in rotated animals. Motion sickness responses were differentially inhibited by orexin-A injection into the DMV (anorexia and hypoactivity), the PVN (conditioned gaping) and the PMV (hypothermia). SB-334867 and TCS-OX2-29 (i.c.v. and intranucleus injection) inhibited behavioural and thermal effects of orexin-A. Orexin-A (60 µg·kg-1) and scopolamine inhibited rotation-induced emesis and non-retching/vomiting symptoms, while orexin-A also attenuated anorexia with mild salivation in motion sickness cats. CONCLUSION AND IMPLICATIONS: Orexin-A might relieve motion sickness through acting on OX1 and OX2 receptors in various hypothalamus nuclei. Intranasal orexin-A could be a potential strategy against motion sickness.


Asunto(s)
Benzoxazoles , Hipotermia , Mareo por Movimiento , Naftiridinas , Urea/análogos & derivados , Ratas , Gatos , Animales , Orexinas/farmacología , Receptores de Orexina/metabolismo , Anorexia/metabolismo , Hipotálamo/metabolismo , Mareo por Movimiento/tratamiento farmacológico , Mareo por Movimiento/metabolismo , Escopolamina/metabolismo , Escopolamina/farmacología , Antagonistas de los Receptores de Orexina/metabolismo , Antagonistas de los Receptores de Orexina/farmacología
3.
J Neurosci Res ; 101(8): 1305-1323, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37012516

RESUMEN

Orexin-A (OXA) is a hypothalamic neuropeptide implicated in the regulation of wakefulness, appetite, reward processing, muscle tone, motor activity, and other physiological processes. The broad range of systems affected stems from the widespread projections of orexin neurons toward multiple brain regions regulating numerous physiological processes. Orexin neurons integrate nutritional, energetic, and behavioral cues and modulate the functions of target structures. Orexin promotes spontaneous physical activity (SPA), and we recently showed that orexin injected into the ventrolateral preoptic area (VLPO) of the hypothalamus increases behavioral arousal and SPA in rats. However, the specific mechanisms underlying the role of orexin in physical activity are unknown. Here we tested the hypothesis that OXA injected into the VLPO alters the oscillatory activity in the electroencephalogram (EEG) to reflect an increased excitability of the sensorimotor cortex, which may explain the associated increase in SPA. The results showed that OXA increased wakefulness following injections into the VLPO. In addition, OXA altered the power spectrum of the EEG during the awake state by decreasing the power of 5-19 Hz oscillations and increasing the power of >35 Hz oscillations, which are markers of increased sensorimotor excitability. Consistently, we found that OXA induced greater muscle activity. Furthermore, we found a similar change in power spectrum during slow-wave sleep, which suggests that OXA altered the EEG activity in a fundamental way, even in the absence of physical activity. These results support the idea that OXA increases the excitability of the sensorimotor system, which may explain the corresponding increase in awake time, muscle tone, and SPA.


Asunto(s)
Tono Muscular , Área Preóptica , Ratas , Animales , Orexinas/farmacología , Orexinas/metabolismo , Área Preóptica/metabolismo , Sueño/fisiología , Hipotálamo/metabolismo , Vigilia/fisiología
4.
Oxid Med Cell Longev ; 2022: 6080282, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211826

RESUMEN

Cognitive impairment (CI) related to Alzheimer's disease (AD) and vascular cognitive disorders (VCDs) has become a key problem worldwide. Importantly, CI is a neuropsychiatric abnormality mainly characterized by learning and memory impairments. The hippocampus is an important brain region controlling learning and memory. Recent studies have highlighted the effects of acupuncture on memory deficits in AD and VCDs. By reviewing the literature published on this topic in the past five years, the present study intends to summarize the effects of acupuncture on memory impairment in AD and VCDs. Focusing on hippocampal synaptic plasticity, we reviewed the mechanisms underlying the effects of acupuncture on memory impairments through regulation of synaptic proteins, AD characteristic proteins, intestinal microbiota, neuroinflammation, microRNA expression, orexin system, energy metabolism, etc., suggesting that hippocampal synaptic plasticity may be the common as well as the core link underlying the above mechanisms. We also discussed the potential strategies to improve the effect of acupuncture. Additionally, the effects of acupuncture on synaptic plasticity through the regulation of vascular-glia-neuron unit were further discussed.


Asunto(s)
Terapia por Acupuntura , Enfermedad de Alzheimer , MicroARNs , Enfermedad de Alzheimer/metabolismo , Animales , Cognición , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Trastornos de la Memoria/metabolismo , MicroARNs/metabolismo , Plasticidad Neuronal , Orexinas/metabolismo , Orexinas/farmacología
5.
J Neurophysiol ; 127(5): 1221-1229, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35353632

RESUMEN

The regulation of sleep/wake behavior and energy homeostasis is maintained in part by the hypothalamic neuropeptide orexin A (OXA, hypocretin). Reduction in orexin signaling is associated with sleep disorders and obesity, whereas higher lateral hypothalamic (LH) orexin signaling and sensitivity promotes obesity resistance. Similarly, dysregulation of hypothalamic neural networks is associated with onset of age-related diseases, including obesity and several neurological diseases. Despite the association of obesity and aging, and that adult populations are the target for the majority of pharmaceutical and obesity studies, conventional models for neuronal networks utilize embryonic neural cultures rather than adult neurons. Synchronous activity describes correlated changes in neuronal activity between neurons and is a feature of normal brain function, and is a measure of functional connectivity and final output from a given neural structure. Earlier studies show alterations in hypothalamic synchronicity following behavioral perturbations in embryonic neurons obtained from obesity-resistant rats and following application of orexin onto embryonic hypothalamic cultures. Synchronous network dynamics in adult hypothalamic neurons remain largely undescribed. To address this, we established an adult rat hypothalamic culture in multi-electrode-array (MEA) dishes and recorded the field potentials. Then we studied the effect of exogenous orexin on network synchronization of these adult hypothalamic cultures. In addition, we studied the wake promoting effects of orexin in vivo when directly injected into the lateral hypothalamus (LH). Our results showed that the adult hypothalamic cultures are viable for nearly 3 mo in vitro, good quality MEA recordings can be obtained from these cultures in vitro, and finally, that cultured adult hypothalamus is responsive to orexin. These results support that adult rat hypothalamic cultures could be used as a model to study the neural mechanisms underlying obesity. In addition, LH administration of OXA enhanced wakefulness in rats, indicating that OXA enhances wakefulness partly by promoting neural synchrony in the hypothalamus.NEW & NOTEWORTHY This study, for the first time, demonstrates that adult hypothalamic cultures are viable in vitro for a prolonged duration and are electrophysiologically active. In addition, the study shows that orexin enhances neural synchronization in adult hypothalamic cultures.


Asunto(s)
Área Hipotalámica Lateral , Hipotálamo , Animales , Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Obesidad , Orexinas/farmacología , Ratas
6.
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
7.
J Psychiatry Neurosci ; 46(4): E459-E471, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34318655

RESUMEN

Background: Orexin-A (OrxA) administration in the posterior paraventricular nucleus of the thalamus (pPVT) reinstates extinguished cocaine-seeking behaviour following extended access to the drug (a model of dependence). The pPVT receives and integrates information associated with emotionally salient events and sends excitatory inputs to brain regions involved in the expression of emotional states, such as those driving cocaine-seeking behaviour (i.e., the nucleus accumbens, the central nucleus of the amygdala [CeA], the basolateral amygdala, the bed nucleus of the stria terminalis [BNST] and the prefrontal cortex). Methods: We monitored the activation pattern of these regions (measured by Fos) during cocaine-seeking induced by OrxA administered to the pPVT. The BNST and CeA emerged as being selectively activated. To test whether the functionality of these regions was pivotal during OrxA-induced cocaine-seeking behaviour, we transiently inactivated these regions concomitantly with OrxA administration to the pPVT. We then tested the participation of corticotropin-releasing factor receptors (CRF1) in the CeA during OrxA-induced cocaine-seeking using the CRF1 antagonist CP154526. Results: We observed selective activation of the CeA and BNST during cocaine-seeking induced by OrxA administered to the pPVT, but only transient inactivation of the CeA prevented cocaine-seeking behaviour. Administration of CP154526 to the CeA prevented OrxAinduced cocaine-seeking behaviour. Limitations: The use of only male rats could have been a limitation. Other limitations could have been the use of an indirect approach to test the hypothesis that administration of OrxA to the pPVT drives cocaine-seeking via CRF1 signalling in the CeA, and a lack of analysis of the participation of CeA subregions. Conclusion: Cocaine-seeking behaviour induced by OrxA administered to the pPVT is driven by activation of the CeA via CRF1 signalling.


Asunto(s)
Núcleo Amigdalino Central/efectos de los fármacos , Trastornos Relacionados con Cocaína/prevención & control , Cocaína , Orexinas/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Tálamo/efectos de los fármacos , Animales , Cocaína/farmacología , Masculino , Orexinas/administración & dosificación , Ratas
8.
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
9.
Pflugers Arch ; 471(11-12): 1407-1418, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31667577

RESUMEN

Orexin A (OXA) is a neuropeptide associated with plasma insulin and leptin levels involved in body weight and appetite regulation. However, little is known about the effect of OXA on leptin secretion in adipocytes and its physiological roles. Leptin secretion and expression were analysed in 3T3-L1 adipocytes. Plasma leptin, adiponectin and insulin levels were measured by ELISA assay. Phosphorylated signal transducer and activator of transcription 3 (pSTAT3) levels in the hypothalamus were evaluated by western blotting. OXA dose-dependently suppressed leptin secretion from 3T3-L1 adipocytes by inhibiting its gene expression while facilitating adiponectin secretion. The leptin inhibition by OXA was mediated via orexin receptors (OXR1 and OXR2). In addition to the pathway via extracellular signal-regulated kinases, OXA triggered adenylyl cyclase-induced cAMP elevation, which results in protein kinase A-mediated activation of cAMP response element-binding proteins (CREB). Accordingly, CREB inhibition restored the OXA-induced downregulation of leptin gene expression and secretion. Exogenous OXA for 4 weeks decreased fasting plasma leptin levels and increased hypothalamic pSTAT3 levels in high-fat diet-fed mice, regardless of increase in body weight and food intake. These results suggest that high dose of OXA directly inhibits leptin mRNA expression and thus secretion in adipocytes, which may be a peripheral mechanism of OXA for its role in appetite drive during fasting. It may be also critical for lowering basal plasma leptin levels and thus maintaining postprandial hypothalamic leptin sensitivity.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/sangre , Leptina/metabolismo , Orexinas/farmacología , Células 3T3-L1 , Animales , Apetito/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Línea Celular , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/metabolismo , Receptores de Orexina/metabolismo
10.
Sci Rep ; 9(1): 8198, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31160650

RESUMEN

Stress is one of major factors that cause sleep problems. Hypocretin represents a stress-related neuropeptide and is well known in maintaining physiological wakefulness. The hypocretinergic neurons originate in the lateral hypothalamic area (LHA) and transmit to several brain regions, including the median raphe nuclei (MRNs). The MRNs modulate both fear responses and sleep-wake activity; however, it remains unclear whether stress alters the levels of hypocretin to regulate MRNs and consequently disrupt sleep. In this paper, we employed the inescapable footshock stimuli (IFS) as a stressor and hypothesized that the IFS-induced sleep disruption is mediated by increased hypocretins in the MRNs. Our results demonstrate that the concentrations of hypocretin in the hypothalamus increased after IFS. Rapid eye movement (REM) sleep was reduced after footshock, and microinjection of non-selective hypocretin receptor antagonist TCS-1102 into the MRNs blocked the IFS-induced decrease of REM sleep. Furthermore, administration of hypocretins into the MRNs mimicked the IFS-induced REM sleep reduction. These results conclude that the increased levels of hypocretins in the MRNs mediate the IFS-induced REM sleep reduction.


Asunto(s)
Mapeo Encefálico/métodos , Orexinas/farmacología , Núcleos del Rafe/fisiología , Sueño REM , Animales , Miedo , Área Hipotalámica Lateral/fisiología , Hipotálamo/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neuropéptidos/metabolismo , Receptores de Orexina , Orexinas/metabolismo , Ratas , Ratas Wistar , Sueño , Estrés Fisiológico , Vigilia
11.
Biochem Biophys Res Commun ; 514(1): 166-172, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31029425

RESUMEN

Orexin is known as an important neuropeptide in the regulation of energy metabolism. However, the role of orexin in exercise-induced leptin sensitivity in the hypothalamus has been unclear. In this study, we determined the effect of transient treadmill exercise on leptin sensitivity in the mediobasal hypothalamus (MBH) of mice and examined the role of orexin in post-exercise leptin sensitivity. Treadmill running for 45 min increased the orexin neuron activity in mice. Intraperitoneal injection of a submaximal dose of leptin after exercise stimulated the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in MBH of mice post-exercise compared with that in non-exercised mice, although intracerebroventricular (icv) injection of leptin did not enhance STAT3 phosphorylation, even after exercise. Icv injection of an orexin receptor antagonist, SB334867 reduced STAT3 phosphorylation, which was enhanced by icv injection of orexin but not by direct injection of orexin into MBH. Exercise increased the phosphorylation of extracellular signal-regulated kinases (ERKs) in the MBH of mice, while ERK phosphorylation was reduced by SB334867. Leptin injection after exercise increased the leptin level in MBH, whereas icv injection of SB334867 suppressed the increase in the leptin level in MBH of mice. These results indicate that the activation of orexin neurons by exercise may contribute to the enhancement of leptin sensitivity in MBH. This effect may be mediated by increased transportation of circulating leptin into MBH, with the involvement of ERK phosphorylation.


Asunto(s)
Hipotálamo/fisiología , Leptina/farmacología , Orexinas/metabolismo , Animales , Benzoxazoles/farmacología , Prueba de Esfuerzo , Hipotálamo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Naftiridinas/farmacología , Neuronas/efectos de los fármacos , Antagonistas de los Receptores de Orexina/farmacología , Receptores de Orexina/metabolismo , Orexinas/farmacología , Fosforilación , Condicionamiento Físico Animal , Receptores de Leptina/metabolismo , Factor de Transcripción STAT3/metabolismo , Urea/análogos & derivados , Urea/farmacología
12.
Mol Psychiatry ; 24(2): 282-293, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30087452

RESUMEN

Hypothalamic neuropeptide orexin has been implicated in the pathophysiology of psychiatric disorders and accumulating clinical evidence indicates a potential link between orexin and depression. However, the exact role of orexin in depression, particularly the underlying neural substrates and mechanisms, remains unknown. In this study, we reveal a direct projection from the hypothalamic orexinergic neurons to the ventral pallidum (VP), a structure that receives an increasing attention for its critical position in rewarding processing, stress responses, and depression. We find that orexin directly excites GABAergic VP neurons and prevents depressive-like behaviors in rats. Two orexin receptors, OX1R and OX2R, and their downstream Na+-Ca2+ exchanger and L-type Ca2+ channel co-mediate the effect of orexin. Furthermore, pharmacological blockade or genetic knockdown of orexin receptors in VP increases depressive-like behaviors in forced swim test and sucrose preference test. Intriguingly, blockage of orexinergic inputs in VP has no impact on social proximity in social interaction test between novel partners, but remarkably strengthens social avoidance under an acute psychosocial stress triggered by social rank. Notably, a significantly increased orexin level in VP is accompanied by an increase in serum corticosterone in animals exposed to acute stresses, including forced swimming, food/water deprivation and social rank stress, rather than non-stress situations. These results suggest that endogenous orexinergic modulation on VP is especially critical for protecting against depressive reactions to stressful events. The findings define an indispensable role for the central orexinergic system in preventing depression by promoting stress resilience.


Asunto(s)
Depresión/tratamiento farmacológico , Orexinas/metabolismo , Orexinas/farmacología , Animales , Conducta Animal/efectos de los fármacos , Depresión/fisiopatología , Modelos Animales de Enfermedad , Neuronas GABAérgicas/metabolismo , Hipotálamo/metabolismo , Masculino , Neuropéptidos/metabolismo , Receptores de Orexina/metabolismo , Receptores de Orexina/fisiología , Ratas , Ratas Sprague-Dawley , Conducta Social , Estrés Psicológico/metabolismo
13.
Biosci Biotechnol Biochem ; 82(12): 2168-2175, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30240332

RESUMEN

We previously reported an orexigenic action of oral zinc administration in male Sprague-Dawley (SD) rats during an early stage of feeding with a zinc-deficient diet, without decreased zinc concentrations in tissues. The overall conclusion was that orally but not intraperitoneally administered zinc stimulates food intake in short-term zinc-deficient-diet fed rats. We here investigate the mechanism of the orexigenic action of zinc using GC-MS/MS-targeted metabolomic analysis in the rat hypothalamus. Four-week-old, male SD/Slc rats were used, and after 2 days of feeding with a zinc-deficient diet, 3 mg of ZnSO4 in 5 mL saline solution were administered to each rat either orally or intraperitoneally. Three hours after administration, the rats were sacrificed and the hypothalamus were excised and analyzed. We found that the oral administration group showed increased concentrations of 3-aminopropanoic acid (ß-alanine), hypotaurine, dopamine, and biotin. In light of metabolomic analysis of these results, we indicate directions for further research.


Asunto(s)
Hipotálamo/metabolismo , Metabolómica , Orexinas/farmacología , Sulfato de Zinc/administración & dosificación , Zinc/deficiencia , Administración Oral , Animales , Apetito/fisiología , Biotina/metabolismo , Cromatografía Líquida de Alta Presión , Dieta , Dopamina/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Inyecciones Intraperitoneales , Masculino , Ratas , Ratas Sprague-Dawley , Taurina/análogos & derivados , Taurina/metabolismo , Sulfato de Zinc/farmacología , beta-Alanina/metabolismo
14.
Neuromolecular Med ; 20(4): 525-536, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30218420

RESUMEN

It is an established fact that orexin plays an important role in regulating the reproductive axis and the secretions of gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH). However, its precise cellular and molecular mechanisms are not fully recognized. Accordingly, the aim of the present study is to find out whether the central injection of orexin A (OXA) and its antagonists, SB-334867 (as orexin receptor antagonist 1; OX1RA) and JNJ-10397049 (as orexin receptor antagonist 2; OX2RA), either alone or in combination, can leave any impact on the reproductive axis (either hormonal or behavioral) in the male Wistar rats. Furthermore, in order to see whether OXA signals can be relayed through the pathway of kisspeptin/neurokinin B/dynorphin (known as KNDy neurons, a neural network which works upstream of GnRH neurons) or not, the relative gene expression of these neuropeptides were measured. Overall, the data from radioimmunoassay revealed that OXA significantly decreases the mean serum level of LH and testosterone and, in a similar vein, its antagonists neutralize this impact. Moreover, data from real-time quantitative PCR indicated that OXA has significantly reduced the hypothalamic expression of Gnrh. In this line, the gene expressions of Kisspeptin and Neurokinin b decreased. However, OXA antagonists neutralize this impact. Also, the expression of Dynorphin gene was upregulated by the following application of the OXA. The results of this study are related to the impact of orexin on the reproductive axis. It is recommended that KNDy neurons as the interneural pathway relay the information of orexin to the GnRH neurons.


Asunto(s)
Dinorfinas/metabolismo , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Neuronas/efectos de los fármacos , Orexinas/farmacología , Reproducción/efectos de los fármacos , Animales , Benzoxazoles/administración & dosificación , Benzoxazoles/farmacología , Dioxanos/administración & dosificación , Dioxanos/farmacología , Dinorfinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Inyecciones Intraventriculares , Kisspeptinas/genética , Hormona Luteinizante/sangre , Masculino , Naftiridinas , Neuroquinina B/genética , Neuronas/metabolismo , Orexinas/administración & dosificación , Orexinas/antagonistas & inhibidores , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Distribución Aleatoria , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Conducta Sexual Animal/efectos de los fármacos , Testosterona/sangre , Urea/administración & dosificación , Urea/análogos & derivados , Urea/farmacología
15.
Biochimie ; 152: 94-109, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29964087

RESUMEN

Orexin A (OXA), a hypothalamic neuropeptide, regulates food intake, sleep-wake cycle and energy balance by binding to its receptor (OX1R). Apart from brain, OXA and OX1R are also present in peripheral organs including reproductive tissues. Mammalian reproduction depends on uptake and proper utilization of glucose in the testes. This study, therefore, examined role of OXA/OX1R system in regulation of glucose homeostasis in adult mouse testis under in vivo and ex vivo conditions. Binding of OXA to OX1R was blocked using an OX1R antagonist, SB-334867. Mice were given a single bilateral intratesticular injection of the antagonist at doses of 4 and 12µg/mouse and sacrificed 24 h post-injection. In order to understand the direct role of OXA in testes of adult mice, an ex vivo experiment was performed where binding of OXA to OX1R in the testis was blocked by using the same OX1R antagonist. The antagonist treatment affected testicular glucose and lactate concentration with concomitant down-regulation in the expression of glucose transporters 3 and 8. A decreased activity in lactate dehydrogenase enzyme and imbalance between germ cell survival and proliferation were also noted in testes in treated mice. The results of ex vivo study supported the results obtained from in vivo study. The findings thus suggest involvement of OXA/OX1R system in regulation of testicular glucose homeostasis and germ cell kinetics in adult mice.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Hipotálamo/metabolismo , Orexinas/fisiología , Espermatozoides/metabolismo , Animales , Benzoxazoles/farmacología , Western Blotting , Ciclo Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Etiquetado Corte-Fin in Situ , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Naftiridinas , Receptores de Orexina/metabolismo , Orexinas/administración & dosificación , Orexinas/metabolismo , Orexinas/farmacología , Espermatozoides/citología , Urea/análogos & derivados , Urea/farmacología
16.
ACS Chem Neurosci ; 9(3): 587-602, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29129052

RESUMEN

Orexins are hypothalamic neuropeptides playing important roles in many functions including the motivation of addictive behaviors. Blockade of the orexin-1 receptor has been suggested as a potential strategy for the treatment of drug addiction. We have previously reported OX1 receptor antagonists based on the tetrahydroisoquinoline scaffold with excellent OX1 potency and selectivity; however, these compounds had high lipophilicity (clogP > 5) and low to moderate solubility. In an effort to improve their properties, we have designed and synthesized a series of analogues where the 7-position substituents known to favor OX1 potency and selectivity were retained, and groups of different nature were introduced at the 1-position where substitution was generally tolerated as demonstrated in previous studies. Compound 44 with lower lipophilicity (clogP = 3.07) displayed excellent OX1 potency ( Ke = 5.7 nM) and selectivity (>1,760-fold over OX2) in calcium mobilization assays. In preliminary ADME studies, 44 showed excellent kinetic solubility (>200 µM), good CNS permeability ( Papp = 14.7 × 10-6 cm/sec in MDCK assay), and low drug efflux (efflux ratio = 3.3).


Asunto(s)
Receptores de Orexina/efectos de los fármacos , Orexinas/farmacología , Permeabilidad/efectos de los fármacos , Tetrahidroisoquinolinas/farmacología , Animales , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Antagonistas de los Receptores de Orexina/farmacología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Solubilidad
17.
Int J Obes (Lond) ; 42(4): 737-745, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29180723

RESUMEN

BACKGROUND/OBJECTIVES: Low levels of orexin are associated with obesity and reduced physical activity in humans and animals. SUBJECTS/METHODS: Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) selectively activated orexin neurons in mouse lateral hypothalamus (LH) to measure effects on spontaneous physical activity (SPA). DREADD targeting was achieved by stereotaxic injection of AAV vectors into caudal lateral LH of heterozygous orexin-Cre or C57/B6J mice. In one set of studies, excitation of orexin neurons was examined (virus: AAV2-EF1a-DIO-hM3Dq-mCherry), and test sessions began 3-4 h after light cycle onset. In a study examining the inhibition of orexin neurons (virus: AAV2-hSyn-DIO-hM4Di-mCherry), testing began 15 min prior to dark cycle onset. Clozapine n-oxide (CNO; 1 or 5 mg/kg) or saline was injected intraperitoneally and time spent moving in open field chambers was recorded for 2 h. Follow-up studies in separate mouse cohorts quantified SPA in parallel with changes in energy expenditure (EE) and chow intake using indirect calorimetry chambers (SableSystem™). Following acclimation, testing sessions (saline and/or CNO) took place over the course of ~1 week, with injections administered every day. Changes in SPA, EE, chow intake, fecal boli, and body composition (EchoMRI™) were measured. Additional mice cohorts were fed a high-fat diet (HFD) and injected with CNO daily up to 10 days to assess the potential for orexin activation to prevent diet-induced obesity. RESULTS: Activation of orexin resulted in increases in SPA in male and female mice, and was accompanied by increases in energy expenditure without changes in overall chow intake. When orexin activation occurred in the context of high fat diet, weight gain and adiposity were significantly attenuated. SPA was decreased when DREADDs were used to inhibit orexin activity. CONCLUSION: These results demonstrate that orexin neurons play a critical role in mediating physical activity and suggest a novel therapeutic target for treating obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Orexinas/metabolismo , Orexinas/farmacología , Adiposidad/fisiología , Animales , Dieta Alta en Grasa , Femenino , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
18.
Sci Rep ; 7: 41535, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28145492

RESUMEN

Hypocretin/Orexin (H/O) neurons of the lateral hypothalamus are compelling modulator candidates for the chronobiology of neuroendocrine output and, as a consequence, hormone release from the anterior pituitary. Here we investigate the effects of H/O peptides upon tuberoinfundibular dopamine (TIDA) neurons - cells which control, via inhibition, the pituitary secretion of prolactin. In whole cell recordings performed in male rat hypothalamic slices, application of H/O-A, as well as H/O-B, excited oscillating TIDA neurons, inducing a reversible depolarising switch from phasic to tonic discharge. The H/O-induced inward current underpinning this effect was post-synaptic (as it endured in the presence of tetrodotoxin), appeared to be carried by a Na+-dependent transient receptor potential-like channel (as it was blocked by 2-APB and was diminished by removal of extracellular Na+), and was a consequence of OX2R receptor activation (as it was blocked by the OX2R receptor antagonist TCS OX2 29, but not the OX1R receptor antagonist SB 334867). Application of the hormone, melatonin, failed to alter TIDA membrane potential or oscillatory activity. This first description of the electrophysiological effects of H/Os upon the TIDA network identifies cellular mechanisms that may contribute to the circadian rhythmicity of prolactin secretion.


Asunto(s)
Cationes/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Receptores de Orexina/metabolismo , Orexinas/farmacología , Potenciales Sinápticos/efectos de los fármacos , Animales , Compuestos de Boro/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Melatonina/metabolismo , Melatonina/farmacología , Sistemas Neurosecretores/efectos de los fármacos , Sistemas Neurosecretores/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Sodio/metabolismo , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
19.
Neuropeptides ; 62: 11-20, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28173961

RESUMEN

In the paraventricular nucleus of the mammalian hypothalamus, hypophysiotropic thyrotropin releasing hormone (TRH) neurons integrate metabolic information and control the activity of the thyroid axis. Additional populations of TRH neurons reside in various hypothalamic areas, with poorly defined connections and functions, albeit there is evidence that some may be related to energy balance. To establish extracellular modulators of TRH hypothalamic neurons activity, we performed a screen of neurotransmitters effects in hypothalamic cultures. Cell culture conditions were chosen to facilitate the full differentiation of the TRH neurons; these conditions had permitted the characterization of the effects of known modulators of hypophysiotropic TRH neurons. The major end-point of the screen was Trh mRNA levels, since they are generally rapidly (0.5-3h) modified by synaptic inputs onto TRH neurons; in some experiments, TRH cell content or release was also analyzed. Various modulators, including histamine, serotonin, ß-endorphin, met-enkephalin, and melanin concentrating hormone, had no effect. Glutamate, as well as ionotropic agonists (kainate and N-Methyl-d-aspartic acid), increased Trh mRNA levels. Baclofen, a GABAB receptor agonist, and dopamine enhanced Trh mRNA levels. An endocannabinoid receptor 1 inverse agonist promoted TRH release. Somatostatin increased Trh mRNA levels and TRH cell content. Orexin-A rapidly increased Trh mRNA levels, TRH cell content and release, while orexin-B decreased Trh mRNA levels. These data reveal unaccounted regulators, which exert potent effects on hypothalamic TRH neurons in vitro.


Asunto(s)
Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Orexinas/farmacología , Hormona Liberadora de Tirotropina/metabolismo , Animales , Células Cultivadas , Hormonas Hipotalámicas/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Hormonas Hipofisarias/metabolismo , Precursores de Proteínas/metabolismo , Ácido Pirrolidona Carboxílico/farmacología , Ratas Wistar , Glándula Tiroides/metabolismo , Tirotropina/metabolismo
20.
PLoS One ; 12(1): e0169908, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28085909

RESUMEN

Orexin-A and -B are hypothalamic neuropeptides of 33 and 28-amino acids, which regulate many homeostatic systems including sleep/wakefulness states, energy balance, energy homeostasis, reward seeking and drug addiction. Orexin-A treatment was also shown to reduce tumor development in xenografted nude mice and is thus a potential treatment for carcinogenesis. The aim of this work was to explore in healthy mice the consequences on energy expenditure components of an orexin-A treatment at a dose previously shown to be efficient to reduce tumor development. Physiological approaches were used to evaluate the effect of orexin-A on food intake pattern, energy metabolism body weight and body adiposity. Modulation of the expression of brain neuropeptides and receptors including NPY, POMC, AgRP, cocaine- and amphetamine related transcript (CART), corticotropin-releasing hormone (CRH) and prepro-orexin (HCRT), and Y2 and Y5 neuropeptide Y, MC4 (melanocortin), OX1 and OX2 orexin receptors (Y2R, Y5R, MC4R, OX1R and OX2R, respectively) was also explored. Our results show that orexin-A treatment does not significantly affect the components of energy expenditure, and glucose metabolism but reduces intraperitoneal fat deposit, adiposity and the expression of several brain neuropeptide receptors suggesting that peripheral orexin-A was able to reach the central nervous system. These findings establish that orexin-A treatment which is known for its activity as an inducer of tumor cell death, do have minor parallel consequence on energy homeostasis control.


Asunto(s)
Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Orexinas/farmacología , Animales , Conducta Alimentaria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos BALB C , Receptores de Orexina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA