Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gastroenterol Hepatol ; 43(5): 240-247, 2020 May.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31983458

RESUMEN

OBJECTIVE: Gastric cancer (GC) has been become the second leading cause for cancer-associated death. This study aimed to investigate Orexin A levels and associated receptors in tumor tissues of GC patients. PATIENTS AND METHODS: Forty-six consecutive gastric cancer patients (GC, n=46) and 13 chronic atrophic gastritis patients (CAG, n=13) were recruited. Meanwhile, 18 health individuals visiting Medical Examination Department were involved as control (N group, n=18). ELISA was used to examine Orexin A concentration. Immunohistochemistry assay was used to examine OX1R and OX2R. HE staining was applied to evaluate inflammation. qRT-PCR was employed to detect OX1R, OX2R, prepro-Orexin mRNAs. Serum Helicobacter pylori (H. pylori) infection was measured. RESULTS: Orexin A expression in GC patients was significantly up-regulated compared to N group and CAG group (p<0.05). Orexin A expression was increased in CAG group compared to N group (p<0.05). Gastric cancer tissues exhibited significantly obvious inflammation compared to N group and CAG group (p<0.05). OX1R and OX2R expressions were significantly down-regulated in GC group compared to N group and CAG group (p<0.05). OX1R and OX2R were lower significantly in GC group compared to CAG group (p<0.05). Prepro-Orexin was significantly depleted in tumor tissues of GC group compared to N group and CAG group (p<0.05). Orexin A expression was un-associated with gender, age and differential grades (p>0.05). CAG and GC patients demonstrated higher H. pylori infection rates. CONCLUSION: Orexin A was associated with inflammation by interacting with OX1R/OX2R receptor and activating prepro-Orexin in tumor tissues of gastric cancer patients.


Asunto(s)
Gastritis/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Proteínas de Neoplasias/fisiología , Receptores de Orexina/fisiología , Orexinas/fisiología , Precursores de Proteínas/metabolismo , Neoplasias Gástricas/metabolismo , Femenino , Gastritis/complicaciones , Gastritis Atrófica/metabolismo , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptores de Orexina/biosíntesis , Receptores de Orexina/genética , Orexinas/biosíntesis , Orexinas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
2.
Naunyn Schmiedebergs Arch Pharmacol ; 392(8): 925-936, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30919010

RESUMEN

We previously reported that hypothalamic tumor necrosis factor-alpha (TNF-α) mRNA expression via histamine H4 receptors contributes to the development of cisplatin-induced anorexia; however, its precise mechanisms remain unclear. It has been reported that chemotherapeutic agents induce the suppression of orexin neuron activity, and the administration of orexin inhibits chemotherapeutic agent-induced gastric discomfort. Other studies demonstrated that the central administration of TNF-α impairs the orexinergic system, and that orexin excites the histaminergic system. We investigated the involvement of orexinergic and histaminergic systems in the therapeutic effect of an H4 receptor antagonist against cisplatin-induced anorexia. Cisplatin decreased the expression of prepro-orexin mRNA, which encodes precursors of orexin, in the hypothalamus of mice. The period of expression decreased in parallel with the onset of anorexia, and treatment with an H4 receptor antagonist (JNJ7777120, 10 mg/kg) inhibited the decrease in expression. The effect of the H4 receptor antagonist on cisplatin-induced anorexia in mice was antagonized by an orexin OX2 receptor antagonist (JNJ10397049, 5 mg/kg) rather than an orexin OX1 receptor antagonist (SB408124, 30 mg/kg). Although an OX2 receptor agonist (YNT-185, 20 mg/kg) or a histamine H3 receptor inverse agonist (ciproxifan, 1 mg/kg) inhibited the cisplatin-induced anorexia, the inhibitory effect of the OX2 receptor agonist was antagonized by an H3 receptor silent antagonist (VUF5681, 5 mg/kg). The combination of JNJ7777120 (10 mg/kg) and ciproxifan (0.5 mg/kg) completely resolved the cisplatin-induced anorexia. These results suggest that activation of the orexinergic and histaminergic pathway is involved in the therapeutic effect of an H4 receptor antagonist against cisplatin-induced anorexia.


Asunto(s)
Anorexia/inducido químicamente , Anorexia/tratamiento farmacológico , Antagonistas de los Receptores Histamínicos/farmacología , Histamina/fisiología , Orexinas/fisiología , Receptores Histamínicos H4/antagonistas & inhibidores , Animales , Anorexia/psicología , Antineoplásicos , Cisplatino , Dioxanos/uso terapéutico , Ingestión de Alimentos/efectos de los fármacos , Agonistas de los Receptores Histamínicos/uso terapéutico , Imidazoles/uso terapéutico , Indoles/uso terapéutico , Masculino , Ratones , Ratones Endogámicos DBA , Receptores de Orexina/efectos de los fármacos , Orexinas/biosíntesis , Compuestos de Fenilurea/uso terapéutico , Piperazinas/uso terapéutico , Transducción de Señal/efectos de los fármacos
3.
Nature ; 566(7744): 383-387, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30760925

RESUMEN

Sleep is integral to life1. Although insufficient or disrupted sleep increases the risk of multiple pathological conditions, including cardiovascular disease2, we know little about the cellular and molecular mechanisms by which sleep maintains cardiovascular health. Here we report that sleep regulates haematopoiesis and protects against atherosclerosis in mice. We show that mice subjected to sleep fragmentation produce more Ly-6Chigh monocytes, develop larger atherosclerotic lesions and produce less hypocretin-a stimulatory and wake-promoting neuropeptide-in the lateral hypothalamus. Hypocretin controls myelopoiesis by restricting the production of CSF1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow. Whereas hypocretin-null and haematopoietic hypocretin-receptor-null mice develop monocytosis and accelerated atherosclerosis, sleep-fragmented mice with either haematopoietic CSF1 deficiency or hypocretin supplementation have reduced numbers of circulating monocytes and smaller atherosclerotic lesions. Together, these results identify a neuro-immune axis that links sleep to haematopoiesis and atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Hematopoyesis/fisiología , Sueño/fisiología , Animales , Antígenos Ly/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células de la Médula Ósea/metabolismo , Femenino , Hematopoyesis/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Mielopoyesis/efectos de los fármacos , Neutrófilos/metabolismo , Receptores de Orexina/deficiencia , Receptores de Orexina/metabolismo , Orexinas/biosíntesis , Orexinas/deficiencia , Orexinas/metabolismo , Orexinas/farmacología , Sueño/efectos de los fármacos , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Privación de Sueño/prevención & control
4.
Sleep ; 42(4)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649483

RESUMEN

STUDY OBJECTIVES: Several studies have reported psychiatric comorbidity in patients with narcolepsy type 1 (NC1). The primary aim of this study was to explore the extent of psychiatric symptoms in a cohort of Norwegian NC1 patients, most of whom were H1N1-vaccinated. We also wanted to explore possible causes of the psychiatric symptoms seen in NC1. METHODS: Cross-sectional study. Psychiatric symptoms were assessed by the Achenbach System of Empirically Based Assessment (ASEBA) Child Behavior Check List (CBCL) in children and by Adult Self Report (ASR) in adults. RESULTS: The mean (SD) total T-scores were 58.6 (9.2) for children and 57.0 (9.8) for adults, these being mainly driven by internalizing problems. Internalizing symptom T-scores showed that 37.5% of the children and 33.3% of the adults were in the clinical range of concern. T-scores were lower when the questionnaire's sleep-related items were excluded. However, 27.5% of children and 22.2% of adults still remained within the total psychiatric symptoms clinical range. Psychiatric symptoms and excessive daytime sleepiness were not associated. However, in children fragmented sleep, measured by sleep-stage shift index was significantly negatively associated with all the psychiatric summary scores (all p ≤ 0.020), and awakening index was negatively associated with externalizing (p = 0.042) and total summary scores (p = 0.042). In adults, awakening index, but not sleep-stage shift index, was positively associated with internalizing score (p = 0.015). Hypocretin-1 levels showed no association with psychiatric symptoms. CONCLUSIONS: We found a high prevalence of psychiatric symptoms in NC1 patients. Fragmented sleep was significantly associated with psychiatric symptoms.


Asunto(s)
Vacunas contra la Influenza/uso terapéutico , Trastornos Mentales/epidemiología , Narcolepsia/epidemiología , Narcolepsia/psicología , Privación de Sueño/psicología , Adolescente , Adulto , Niño , Estudios de Cohortes , Estudios Transversales , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Masculino , Trastornos Mentales/psicología , Noruega/epidemiología , Orexinas/biosíntesis , Privación de Sueño/fisiopatología , Encuestas y Cuestionarios
5.
J Comp Neurol ; 527(9): 1508-1526, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30666646

RESUMEN

Orexin A (OXA) and neuropeptide Y (NPY) are two hypothalamic neuropeptides involved in the regulation of feeding behavior and food intake in all vertebrates. Accumulating evidences document that they undergo age-related modifications, with consequences on metabolism, sleep/wake disorders and progression of neurodegenerations. The present study addressed the age related changes in expression and distribution of orexin A (its precursor is also known as hypocretin-HCRT) and NPY, and their regulation by food intake in the short-lived vertebrate model Nothobranchius furzeri. Our experiments, conducted on male specimens, show that: (a) HCRT and OXA and NPY mRNA and protein are localized in neurons of diencephalon and optic tectum, as well as in numerous fibers projecting through the entire neuroaxis, and are colocalized in specific nuclei; (b) in course of aging, HCRT and NPY expressing neurons are localized also in telencephalon and rhombencephalon; (c) HCRT expressing neurons increased slightly in the diencephalic area of old animals and in fasted animals, whereas NPY increased sharply; (d) central HCRT levels are not regulated neither in course of aging nor by food intake; and (e) central NPY levels are augmented in course of aging, and regulated by food intake only in young. These findings represent a great novelty in the study of central orexinergic and NPY-ergic systems in vertebrates', demonstrating an uncommon and unprecedented described regulation of these two orexigenic neuropeptides.


Asunto(s)
Envejecimiento/metabolismo , Diencéfalo/metabolismo , Ingestión de Alimentos/fisiología , Fundulidae/metabolismo , Regulación de la Expresión Génica/fisiología , Hipotálamo/metabolismo , Neuropéptido Y/biosíntesis , Orexinas/biosíntesis , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Ayuno/metabolismo , Fundulidae/genética , Hibridación in Situ , Masculino , Neuronas/metabolismo , Neuropéptido Y/genética , Orexinas/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Colículos Superiores/metabolismo
6.
J Comp Neurol ; 526(18): 2937-2954, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30019757

RESUMEN

Neurons in the lateral hypothalamic area that express hypocretin (Hcrt) neuropeptides help regulate many behaviors including wakefulness and reward seeking. These neurons project throughout the brain, including to neural populations that regulate wakefulness, such as the locus coeruleus (LC) and tuberomammilary nucleus (TMN), as well as to populations that regulate reward, such as the nucleus accumbens (NAc) and ventral tegmental area (VTA). To address the roles of Hcrt neurons in seemingly disparate behaviors, it has been proposed that Hcrt neurons can be anatomically subdivided into at least two distinct subpopulations: a "medial group" that projects to the LC and TMN, and a "lateral group" that projects to the NAc and VTA. Here, we use a dual retrograde tracer strategy to test the hypotheses that Hcrt neurons can be classified based on their downstream projections and medial/lateral location within the hypothalamus. We found that individual Hcrt neurons were significantly more likely to project to both the LC and TMN or to both the VTA and NAc than would be predicted by chance. In contrast, we found that Hcrt neurons that projected to the LC or TMN were mostly distinct from Hcrt neurons that projected to the VTA or NAc. Interestingly, these two populations of Hcrt neurons are intermingled within the hypothalamus and cannot be classified into medial or lateral groups. These results suggest that Hcrt neurons can be distinguished based on their downstream projections but are intermingled within the hypothalamus.


Asunto(s)
Hipotálamo/citología , Vías Nerviosas/citología , Neuronas/citología , Animales , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Orexinas/análisis , Orexinas/biosíntesis
7.
Pharmacol Rep ; 70(4): 650-657, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29902669

RESUMEN

BACKGROUND: The endocannabinoid (eCB) system is strongly involved in the regulation of anxiety and feeding behavior. RVD-hemopressin(α) [RVD-hp(α)], a N-terminally extended form of hemopressin, is a negative allosteric modulator of the cannabinoid (CB) 1 receptor and a positive allosteric modulator of CB2 receptor which has been recently reported to exert anxiolytic/antidepressant and anorexigenic effects after peripheral administration in rats. Pharmacological evidences reported a possible link between brain hypocretin/orexin, monoamine and eCB systems, as regards appetite and emotional behavior control. Considering this, the aim of our work was to investigated the effects of RVD-hp(α) on anxiety like behavior and food intake after central administration and related it to monoamine levels and orexin-A gene expression, in the hypothalamus. METHODS: We have studied the effects of central RVD-hp(α) (10nmol) injection on anxiety-like behavior and feeding using different behavioral tests. Hypothalamic levels of norepinephrine (NE), dopamine (DA) and serotonin (5-hydroxytryptamine, 5-HT) and gene expression of orexin-A and proopiomelanocortin (POMC) were measured by high performance liquid chromatography (HPLC) and real-time reverse transcription polymerase chain reaction (RT-PCR) analysis, respectively. RESULTS: Central RVD-hp(α) administration decreased locomotion activity and stereotypies. Moreover, RVD-hp(α) treatment inhibited anxiogenic-like behavior and food intake, NE levels and orexin-A gene expression, in the hypothalamus. CONCLUSION: Concluding, in the present study we demonstrated that central RVD-hp(α) induced anxiolytic and anorexigenic effects possibly related to reduced NE and orexin-A and POMC signaling, in the hypothalamus. These findings further support the central role of the peptide in rat brain thus representing an innovative pharmacological approach for designing new anorexigenic drugs targeting eCB system.


Asunto(s)
Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Hemoglobinas/farmacología , Hipotálamo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Norepinefrina/metabolismo , Orexinas/biosíntesis , Fragmentos de Péptidos/farmacología , Animales , Dopamina/metabolismo , Hemoglobinas/administración & dosificación , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Masculino , Actividad Motora/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Proopiomelanocortina/biosíntesis , Ratas , Serotonina/metabolismo , Conducta Estereotipada/efectos de los fármacos
8.
Sci Transl Med ; 10(447)2018 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-29950444

RESUMEN

The changes in brain function that perpetuate opiate addiction are unclear. In our studies of human narcolepsy, a disease caused by loss of immunohistochemically detected hypocretin (orexin) neurons, we encountered a control brain (from an apparently neurologically normal individual) with 50% more hypocretin neurons than other control human brains that we had studied. We discovered that this individual was a heroin addict. Studying five postmortem brains from heroin addicts, we report that the brain tissue had, on average, 54% more immunohistochemically detected neurons producing hypocretin than did control brains from neurologically normal subjects. Similar increases in hypocretin-producing cells could be induced in wild-type mice by long-term (but not short-term) administration of morphine. The increased number of detected hypocretin neurons was not due to neurogenesis and outlasted morphine administration by several weeks. The number of neurons containing melanin-concentrating hormone, which are in the same hypothalamic region as hypocretin-producing cells, did not change in response to morphine administration. Morphine administration restored the population of detected hypocretin cells to normal numbers in transgenic mice in which these neurons had been partially depleted. Morphine administration also decreased cataplexy in mice made narcoleptic by the depletion of hypocretin neurons. These findings suggest that opiate agonists may have a role in the treatment of narcolepsy, a disorder caused by hypocretin neuron loss, and that increased numbers of hypocretin-producing cells may play a role in maintaining opiate addiction.


Asunto(s)
Encéfalo/metabolismo , Cataplejía/tratamiento farmacológico , Narcolepsia/tratamiento farmacológico , Alcaloides Opiáceos/uso terapéutico , Orexinas/biosíntesis , Animales , Encéfalo/patología , Cataplejía/complicaciones , Recuento de Células , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Heroína , Humanos , Masculino , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Morfina/farmacología , Morfina/uso terapéutico , Narcolepsia/complicaciones , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Alcaloides Opiáceos/farmacología , Ratas Sprague-Dawley , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/patología
9.
PLoS One ; 12(6): e0178573, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28570646

RESUMEN

The loss of specific neuronal phenotypes, as determined by immunohistochemistry, has become a powerful tool for identifying the nature and cause of neurological diseases. Here we show that the number of neurons identified and quantified using this method misses a substantial percentage of extant neurons in a phenotype specific manner. In mice, 24% more hypocretin/orexin (Hcrt) neurons are seen in the night compared to the day, and an additional 17% are seen after inhibiting microtubule polymerization with colchicine. We see no such difference between the number of MCH (melanin concentrating hormone) neurons in dark, light or colchicine conditions, despite MCH and Hcrt both being hypothalamic peptide transmitters. Although the size of Hcrt neurons did not differ between light and dark, the size of MCH neurons was increased by 15% in the light phase. The number of neurons containing histidine decarboxylase (HDC), the histamine synthesizing enzyme, was 34% greater in the dark than in the light, but, like Hcrt, cell size did not differ. We did not find a significant difference in the number or the size of neurons expressing choline acetyltransferase (ChAT), the acetylcholine synthesizing enzyme, in the horizontal diagonal band (HBD) during the dark and light conditions. As expected, colchicine treatment did not increase the number of these neurons. Understanding the function and dynamics of transmitter production within "non-visible" phenotypically defined cells has fundamental implications for our understanding of brain plasticity.


Asunto(s)
Ritmo Circadiano , Histamina/biosíntesis , Neuronas/metabolismo , Orexinas/biosíntesis , Animales , Colina O-Acetiltransferasa/metabolismo , Colchicina/administración & dosificación , Histidina Descarboxilasa/metabolismo , Hormonas Hipotalámicas/metabolismo , Masculino , Melaninas/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/enzimología , Hormonas Hipofisarias/metabolismo
10.
J Neurophysiol ; 116(5): 2250-2259, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559138

RESUMEN

Evidence of impaired function of orexin neurons has been found in individuals with cardiorespiratory disorders, such as obstructive sleep apnea (OSA) and sudden infant death syndrome (SIDS), but the mechanisms responsible are unknown. Individuals with OSA and SIDS experience repetitive breathing cessations and/or rebreathing of expired air, resulting in hypoxia/hypercapnia (H/H). In this study, we examined the responses of fluorescently identified rat orexin neurons in the lateral hypothalamus to acute H/H to test if and how these neurons alter their activity and function during this challenge. Experiments were conducted in an in vitro slice preparation using voltage-clamp and current-clamp configurations. H/H (10 min) induced hyperpolarization, accompanied by rapid depression, and finally, cessation of firing activity in orexin neurons. Hypoxia alone had similar but less potent effects. H/H did not alter the frequency of inhibitory glycinergic postsynaptic currents. The frequency of GABAergic currents was diminished but only at 8-10 min of H/H. In contrast, the frequency of excitatory glutamatergic postsynaptic events was diminished as early as 2-4 min of H/H. In the presence of glutamatergic receptor blockers, the inhibitory effects of H/H on the firing activity and membrane potential of orexin neurons persisted but to a lesser extent. In conclusion, both direct alteration of postsynaptic membrane properties and diminished glutamatergic neurotransmission likely contribute to the inhibition of orexin neurons by H/H. These mechanisms could be responsible for the decreased function of orexin in individuals at risk for OSA and SIDS.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Receptores de Orexina/biosíntesis , Consumo de Oxígeno/fisiología , Animales , Hipoxia de la Célula/fisiología , Hipercapnia/metabolismo , Hipotálamo/química , Potenciales de la Membrana/fisiología , Neuronas/química , Receptores de Orexina/análisis , Orexinas/análisis , Orexinas/antagonistas & inhibidores , Orexinas/biosíntesis , Técnicas de Cultivo de Órganos , Ratas , Ratas Transgénicas
11.
Neuropharmacology ; 110(Pt A): 15-24, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27395784

RESUMEN

Stress is a trigger factor for seizure initiation which activates hypothalamic pituitary adrenal (HPA) axis as well other brain areas. In this respect, corticotropin releasing hormone (CRH) and lateral hypothalamus (LH) orexinergic system are involved in seizure occurrence. In this study, we investigated the role of LH area and orexin expression in (mediation of) stress effect on pentylenetetrazol (PTZ) -induced seizures with hippocampal involvement. Two mild foot shock stresses were applied to intact and adrenalectomized animals; with or without CRHr1 blocking (NBI 27914) in the LH area. Then, changes in orexin production were evaluated by RT-PCR. Intravenous PTZ infusion (25 mg/ml) -induced convulsions were scored upon modified Racine scale. Finally, hippocampal glutamate and GABA were evaluated to study excitability changes. We demonstrated that the duration and severity of convulsions in stress-induced as well as adrenalectomized group were increased. Plasma corticosterone (CRT) level and orexin mRNA expression were built up in the stress and/or seizure groups. Furthermore, glutamate and GABA content was increased and decreased respectively due to stress and seizures. In contrast, rats receiving CRHr1 inhibitor showed reduced severity and duration of seizures, increased GABA, decreased glutamate and corticosterone and also orexin mRNA compared to the inhibitor free rats. Stress and adrenalectomy induced augmenting effect on seizure severity and duration and the subsequent reduction due to CRHr1 blocking with parallel orexin mRNA changes, indicated the likely involvement of CRH1r induced orexin expression of the LH in gating stress effect on convulsions.


Asunto(s)
Área Hipotalámica Lateral/metabolismo , Orexinas/biosíntesis , Pentilenotetrazol/toxicidad , Receptores de Hormona Liberadora de Corticotropina/biosíntesis , Convulsiones/metabolismo , Estrés Psicológico/metabolismo , Compuestos de Anilina/farmacología , Animales , Área Hipotalámica Lateral/efectos de los fármacos , Masculino , Pirimidinas/farmacología , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Convulsiones/inducido químicamente , Convulsiones/psicología , Estrés Psicológico/prevención & control , Estrés Psicológico/psicología
12.
Psychoneuroendocrinology ; 67: 171-81, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26907996

RESUMEN

We hypothesize that anorexia nervosa (AN) poses a physiological stress. Therefore, the way an individual copes with stress may affect AN vulnerability. Since prenatal stress (PNS) exposure alters stress responsivity in offspring this may increase their risk of developing AN. We tested this hypothesis using the activity based anorexia (ABA) rat model in control and PNS rats that were characterized by either proactive or passive stress-coping behavior. We found that PNS passively coping rats ate less and lost more weight during the ABA paradigm. Exposure to ABA resulted in higher baseline corticosterone and lower insulin levels in all groups. However, leptin levels were only decreased in rats with a proactive stress-coping style. Similarly, ghrelin levels were increased only in proactively coping ABA rats. Neuropeptide Y (Npy) expression was increased and proopiomelanocortin (Pomc) expression was decreased in all rats exposed to ABA. In contrast, agouti-related peptide (Agrp) and orexin (Hctr) expression were increased in all but the PNS passively coping ABA rats. Furthermore, DNA methylation of the orexin gene was increased after ABA in proactive coping rats and not in passive coping rats. Overall our study suggests that passive PNS rats have innate impairments in leptin and ghrelin in responses to starvation combined with prenatal stress associated impairments in Agrp and orexin expression in response to starvation. These impairments may underlie decreased food intake and associated heightened body weight loss during ABA in the passively coping PNS rats.


Asunto(s)
Adaptación Psicológica/fisiología , Proteína Relacionada con Agouti/biosíntesis , Anorexia/metabolismo , Anorexia/fisiopatología , Orexinas/biosíntesis , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Peso Corporal/fisiología , Metilación de ADN , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Femenino , Ghrelina/biosíntesis , Leptina/biosíntesis , Masculino , Actividad Motora/fisiología , Neuropéptido Y/biosíntesis , Embarazo , Proopiomelanocortina/biosíntesis , Ratas , Regulación hacia Arriba
13.
Folia Histochem Cytobiol ; 53(4): 333-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26714447

RESUMEN

INTRODUCTION: Studies on expression of orexins (OXs) and their receptors in human prostate gland and human prostatic cell lines are scanty and their results contradictory. Regarding this, we carefully reinvestigated this problem on human prostatic cell lines. MATERIAL AND METHODS: Expression of preproorexin (ppOX) (6 primer pairs), and orexin receptors 1 and 2 (OXR1, OXR2) (4 and 2 primer pairs, respectively) was assessed by conventional PCR and QPCR in human normal (PrEC, PrSc, PrSmC) and prostate carcinoma (Du145, LNCaP, and PC3) cell lines. We designed intron spanning primers and also we applied primers from earlier publications and commercially available ones. RESULTS: With the designed primer pairs, in all studied cell lines we failed to demonstrate expression of ppOX, OXR1 and OXR2 genes at the mRNA level, while reaction products were observed in control tissues (human placenta and adrenals). Primers applied in earlier studies did not form amplification products specific for preproorexin or orexin 1 receptor. Some commercially available primers for orexin receptor 1 produced false positive results. CONCLUSIONS: We found no evidence for the presence of preproorexin-orexin receptors system genes' mRNAs in human prostate cell lines. The reported premises for these genes' expression in prostate and prostatic cell lines may have arisen either from the presence of non-prostate cells included in the samples or from faulty PCR settings.


Asunto(s)
Expresión Génica , Receptores de Orexina/biosíntesis , Orexinas/biosíntesis , Neoplasias de la Próstata/metabolismo , Línea Celular , Línea Celular Tumoral , Cartilla de ADN , Humanos , Masculino , Receptores de Orexina/genética , Orexinas/genética , Reacción en Cadena de la Polimerasa , Próstata/metabolismo , Neoplasias de la Próstata/genética
14.
J Mol Endocrinol ; 55(2): 95-106, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26162607

RESUMEN

Neuromedin U (NMU) and neuromedin S (NMS) play inhibitory roles in the regulation of food intake and energy homeostasis in mammals. However, their functions are not clearly established in teleost fish. In the present study, nmu and nms homologs were identified in several fish species. Subsequently, their cDNA sequences were cloned from the orange-spotted grouper (Epinephelus coioides). Sequence analysis showed that the orange-spotted grouper Nmu proprotein contains a 21-amino acid mature Nmu peptide (Nmu-21). The Nms proprotein lost the typical mature Nms peptide, but it retains a putative 34-amino acid peptide (Nmsrp). In situ hybridization revealed that nmu- and nms-expressing cells are mainly localized in the hypothalamic regions associated with appetite regulation. Food deprivation decreased the hypothalamic nmu mRNA levels but induced an increase of nms mRNA levels. Periprandial expression analysis showed that hypothalamic expression of nmu increased significantly at 3 h post-feeding, while nms expression was elevated at the normal feeding time. I.p. injection of synthetic Nmu-21 peptide suppressed the hypothalamic neuropeptide y (npy) expression, while Nmsrp administration significantly increased the expression of npy and orexin in orange-spotted grouper. Furthermore, the mRNA levels of LH beta subunit (lhß) and gh in the pituitary were significantly down-regulated after Nmu-21 peptide administration, while Nmsrp was able to significantly stimulate the expression of FSH beta subunit (fshß), prolactin (prl), and somatolaction (sl). Our results indicate that nmu and nms possess distinct neuroendocrine functions and pituitary functions in the orange spotted grouper.


Asunto(s)
Lubina/genética , Metabolismo Energético/genética , Proteínas de Peces/genética , Neuropéptido Y/genética , Neuropéptidos/genética , Secuencia de Aminoácidos , Animales , Apetito/genética , Secuencia de Bases , Clonación Molecular , Ingestión de Alimentos/genética , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Hipotálamo/citología , Hipotálamo/metabolismo , Hibridación in Situ , Hormona Luteinizante de Subunidad beta/genética , Datos de Secuencia Molecular , Neuropéptido Y/biosíntesis , Neuropéptidos/biosíntesis , Orexinas/biosíntesis , Hipófisis/metabolismo , Prolactina/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Análisis de Secuencia de ADN , Inanición/genética
15.
J Neurotrauma ; 32(5): 289-96, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25242371

RESUMEN

Traumatic brain injury (TBI) can cause sleep-wake disturbances and excessive daytime sleepiness. The pathobiology of sleep disorders in TBI, however, is not well understood, and animal models have been underused in studying such changes and potential underlying mechanisms. We used the rat lateral fluid percussion (LFP) model to analyze sleep-wake patterns as a function of time after injury. Rapid-eye movement (REM) sleep, non-REM (NREM) sleep, and wake bouts during light and dark phases were measured with electroencephalography and electromyography at an early as well as chronic time points after LFP. Moderate TBI caused disturbances in the ability to maintain consolidated wake bouts during the active phase and chronic loss of wakefulness. Further, TBI resulted in cognitive impairments and depressive-like symptoms, and reduced the number of orexin-A-positive neurons in the lateral hypothalamus.


Asunto(s)
Lesiones Encefálicas/complicaciones , Trastornos del Sueño del Ritmo Circadiano/etiología , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Vigilia/fisiología , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Electroencefalografía , Electromiografía , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Orexinas/análisis , Orexinas/biosíntesis , Ratas , Ratas Sprague-Dawley , Trastornos del Sueño del Ritmo Circadiano/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...