Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Gastroenterol Hepatol ; 43(5): 240-247, 2020 May.
Article in English, Spanish | MEDLINE | ID: mdl-31983458

ABSTRACT

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.


Subject(s)
Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Neoplasm Proteins/physiology , Orexin Receptors/physiology , Orexins/physiology , Protein Precursors/metabolism , Stomach Neoplasms/metabolism , Female , Gastritis/complications , Gastritis, Atrophic/metabolism , Gene Expression Regulation, Neoplastic , Helicobacter Infections/complications , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Orexin Receptors/biosynthesis , Orexin Receptors/genetics , Orexins/biosynthesis , Orexins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
2.
J Neurochem ; 147(3): 380-394, 2018 11.
Article in English | MEDLINE | ID: mdl-30102759

ABSTRACT

Orexin is a member of neuropeptides which is involved in the central motor control. The substantia nigra pars compacta (SNc) is an important nucleus participating in motor control under both physiological and pathological conditions. Morphological studies reveal that orexinergic neurons located in lateral hypothalamus innervate the SNc. Both orexin-1 receptors (OX1 R) and orexin-2 receptors (OX2 R) are expressed in the SNc. To investigate the effects of orexins on SNc, single unit in vivo extracellular recordings and behavioral tests were performed in this study. Micro-pressure administration of orexin A and orexin B significantly increased the spontaneous firing rate of nigral DAergic neurons by 65.87 ± 7.73% and 90.49 ± 17.83%, respectively. The excitatory effects of orexin A on nigral DAergic neurons were mainly mediated by OX1 R, while OX2 R were involved in the increase in firing rate induced by orexin B. Selectively blocking OX1 R and OX2 R significantly decreased the firing rate of nigral DAergic neurons by 36.77 ± 6.26% and 32.04 ± 6.12%, respectively, which suggested that endogenous orexins modulated the spontaneous firing activity of nigral DAergic neurons. Finally, both elevated body swing test and haloperidol-induced postural behavioral test showed that unilateral microinjection of orexin A and orexin B induced significantly contralateral-biased swing and deflection behavior. Meanwhile, the specific OX1 R and OX2 R antagonists produced opposite effects. The present electrophysiological and behavioral studies suggested that orexins increased the firing activity of nigral DAergic neurons and participated in central motor control. Open Practices Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Dopaminergic Neurons/drug effects , Orexins/pharmacology , Substantia Nigra/drug effects , Animals , Behavior, Animal , Dopamine Antagonists/pharmacology , Electrophysiological Phenomena/drug effects , Haloperidol/pharmacology , Male , Microinjections , Motor Activity/drug effects , Orexin Receptors/biosynthesis , Orexin Receptors/genetics , Orexins/administration & dosage , Postural Balance/drug effects , Rats , Rats, Wistar , Substantia Nigra/cytology
3.
Neurotoxicology ; 67: 46-53, 2018 07.
Article in English | MEDLINE | ID: mdl-29673962

ABSTRACT

Food intake ensures energy resources sufficient for basic metabolism, immune system and reproductive investment. It is already known that food-seeking performances, which are crucially controlled by orexins (ORXs), may be under the influence of environmental factors including pollutants. Among these, mancozeb (mz) is becoming an environmental risk for neurodegenerative diseases. Due to few studies on marine fish exposed to mz, it was our intention to correlate feeding latency, food intake and feeding duration to potential neurodegenerative processes in key diencephalic sites and expression changes of the ORX neuroreceptor (ORXR) in the ornate wrasses (Thalassoma pavo). Hence, fish exposed for 4 days (d) to mz 0.2 mg/l (deriving from a 0.07, 0.14, 0.2, 0.3 mg/l screening test) displayed a significant reduction (p < 0.05) of food intake compared to controls as early as 1d that became more evident (p < 0.01) after 3d. Moreover, significant enhancements of feeding latency were reported after 1d up to 3d (p < 0.001) and even feeding duration was enhanced up to 3d (p < 0.001), which instead moderately increased after 4d (p < 0.05). A reduction (-120%; p < 0.001) of mean body weight was also detected at the end of exposure. Likewise, a notable (p < 0.001) activation of ORXR protein occurred together with mRNA up-regulations in diencephalic areas such as the diffuse nucleus of the inferior lobe (+48%) that also exhibited evident degenerative neuronal fields. Overall, these results highlight an ORX role as a vital component of the neuroprotective program under environmental conditions that interfere with feeding behaviors.


Subject(s)
Feeding Behavior/drug effects , Fungicides, Industrial/toxicity , Maneb/toxicity , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Orexin Receptors/biosynthesis , Zineb/toxicity , Animals , Feeding Behavior/physiology , Female , Fishes , Gene Expression , Neurodegenerative Diseases/pathology , Orexin Receptors/genetics , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology
4.
Peptides ; 99: 117-127, 2018 01.
Article in English | MEDLINE | ID: mdl-28442349

ABSTRACT

Orexins A/B derived from hypothalamic prepro-orexin (PPO) are agonists for orexin receptors 1 (OX1) and 2 (OX2). Previously, we showed clear sex differences in the hypothalamic-pituitary-gonadal orexinergic system in adult rodents. Here, we studied the effect of sexual brain differentiation on the orexinergic system in neuroendocrine structures regulating reproduction. We evaluated: a: proestrous and neonatally androgenized female rats; b: adult males, untreated or gonadectomized in adulthood and injected with oil or estradiol and progesterone (E2/P4); c: control and demasculinized males (perinatally treated with flutamide and later castration) injected either with oil or E2/P4 in adulthood. Rats were sacrificed at 12:00 and 18:00h; blood samples and brains were collected. Hormones were measured using radioimmunoassay. PPO, OX1 and OX2 mRNAs were quantified by qPCR in medial basal hypothalamus, anterior hypothalamus, adenohypophysis, and cortex. Western blots for OX1 were done in the same structures. In normal females, gonadotropins surged at 18:00h coinciding with significant elevations of PPO, OX1 and OX2 mRNAs and OX1 protein in hypothalamus and pituitary; no increases were observed at noon. Afternoon changes were absent in masculinized females. Demasculinized males when treated with E2/P4 showed high PPO, OX1 and OX2 mRNAs and OX1 protein expression in hypothalamus and pituitary at 12:00 and 18:00h compared vehicle-treated controls. The same steroid treatment was ineffective in males with normal brain masculinization. Here we show that neonatal testosterone shapes the sexual differences in the hypothalamic-pituitary orexinergic system in synchronicity to establishing the brain sex differences of the reproductive axis. The female brain controls gonadotropin surges and concurrent elevations of all studied components of the orexinergic system, suggesting its participation as a possible link between food intake, behavior and hormonal control of reproduction.


Subject(s)
Hypothalamus/metabolism , Orexin Receptors/biosynthesis , Orexins/metabolism , Pituitary Gland, Anterior/metabolism , Sex Characteristics , Testosterone/metabolism , Animals , Estradiol/metabolism , Female , Male , Progesterone/metabolism , Rats , Rats, Sprague-Dawley
5.
Peptides ; 101: 25-31, 2018 03.
Article in English | MEDLINE | ID: mdl-29269074

ABSTRACT

Orexinergic system is involved in reward processing and drug addiction. Objectives here, we investigated the effect of intra-hippocampal CA1 administration of orexin-1 receptor (OX1r) antagonist on the expression, and extinction of morphine-induced place preference in rats. Conditioned place preference (CPP) was induced by subcutaneous injection of morphine (5 mg/kg) during a 3-day conditioning phase. Two experimental plots were designed; SB334867 as a selective OX1r antagonist was dissolved in 12% DMSO, prepared in solutions with different concentrations (3, 30, and 300 nM), and microinjected into the CA1 and some neighboring regions (0.5 µl/side), bilaterally. CPP score and locomotor activity were recorded during the CPP test. Results demonstrated that intra-CA1 administration of the OX1r antagonist attenuates the expression of morphine-induced CPP. Furthermore, higher concentrations of SB334867 facilitated the extinction period of morphine-induced CPP and reduced its latency. Nevertheless, solely administration of DMSO did not have any influence on the CPP scores and locomotion in both phases. Our findings suggest that OX1rs in the CA1 region of the hippocampus are involved in the expression of morphine CPP. Moreover, blockade of OX1rs could facilitate extinction and may extinguish the ability of drug-related cues. It seems that the antagonist might be considered as a propitious therapeutic agent in suppressing drug-seeking behaviors.


Subject(s)
Behavior, Animal/drug effects , CA1 Region, Hippocampal/metabolism , Gene Expression Regulation/drug effects , Morphine/pharmacology , Orexin Receptors/biosynthesis , Animals , Benzoxazoles/pharmacology , Male , Morphine/adverse effects , Naphthyridines , Rats , Rats, Wistar , Urea/analogs & derivatives , Urea/pharmacology
6.
J Neurochem ; 141(2): 222-235, 2017 04.
Article in English | MEDLINE | ID: mdl-28164283

ABSTRACT

Peripheral inflammation is known to trigger neuroinflammation and neurodegenerative disease. However, the key components during the propagation of inflammation from the periphery to the central nervous system (CNS) remain unclear. Lipopolysaccharide (LPS) was administered to Sprague-Dawley rats to induce peripheral inflammation. An intravenous injection and an intranigral injection of clodronate liposomes were given to deplete monocytes and microglia, respectively. Recombinant CD200 fusion protein (CD200Fc) or an anti-CD200R1 antibody was injected into the substantia nigra to manipulate the involvement of CD200 and CD200R1. Immunohistochemistry and immunofluorescence staining were used to measure microglial activation and dopaminergic neuronal loss. The expression of brain pro-inflammatory cytokines (i.e., tumor necrosis factor alpha, IL-1ß) and CD200-CD200R1 signaling were measured by quantitative RT-PCR. Our data showed that the peripheral LPS injection activated the microglia and induced an increase in the levels of pro-inflammatory cytokines (i.e., tumor necrosis factor alpha, IL-1ß). The depletion of either monocytes or microglia suppressed these inflammatory effects that were induced by peripheral LPS administration. The peripheral LPS injection increased the expression of CD200 and CD200R1 in the substantia nigra. Dopaminergic neuronal loss induced by the peripheral LPS injection was accelerated by the blockade of CD200-CD200R1 signaling with an anti-CD200R1 antibody and attenuated by intensifying the signaling with CD200Fc. These results highlight the importance of monocytes, microglia, and CD200-CD200R1 signaling in the transmission of inflammation from the periphery to the CNS.


Subject(s)
Antigens, CD/biosynthesis , Inflammation Mediators/metabolism , Microglia/metabolism , Monocytes/metabolism , Orexin Receptors/biosynthesis , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Inflammation/metabolism , Inflammation/pathology , Male , Microglia/pathology , Monocytes/pathology , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
7.
J Neurophysiol ; 116(5): 2250-2259, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27559138

ABSTRACT

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.


Subject(s)
Hypothalamus/metabolism , Neurons/metabolism , Orexin Receptors/biosynthesis , Oxygen Consumption/physiology , Animals , Cell Hypoxia/physiology , Hypercapnia/metabolism , Hypothalamus/chemistry , Membrane Potentials/physiology , Neurons/chemistry , Orexin Receptors/analysis , Orexins/analysis , Orexins/antagonists & inhibitors , Orexins/biosynthesis , Organ Culture Techniques , Rats , Rats, Transgenic
8.
Folia Histochem Cytobiol ; 53(4): 333-41, 2015.
Article in English | MEDLINE | ID: mdl-26714447

ABSTRACT

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.


Subject(s)
Gene Expression , Orexin Receptors/biosynthesis , Orexins/biosynthesis , Prostatic Neoplasms/metabolism , Cell Line , Cell Line, Tumor , DNA Primers , Humans , Male , Orexin Receptors/genetics , Orexins/genetics , Polymerase Chain Reaction , Prostate/metabolism , Prostatic Neoplasms/genetics
9.
J Chem Neuroanat ; 66-67: 1-9, 2015.
Article in English | MEDLINE | ID: mdl-25841630

ABSTRACT

We have utilised a transgenic reporter mouse in which green fluorescent protein (GFP) expression is driven by the orexin-1 receptor (OX1R) promoter to systematically map the distribution of OX1R-expressing neurons throughout the mouse forebrain and rostral brainstem. GFP labelling was observed in perikarya and fibres in an extensive range of brain loci encompassing the olfactory and cerebral cortices, dorsal and ventral pallidum, hippocampus, amygdaloid regions, septal areas, thalamic nuclei, hypothalamic nuclei and several brainstem regions, consistent with previous studies of OX1R mRNA expression. This is the first study to systematically characterise the neuroanatomical distribution of OX1R in the OX1R-eGFP mouse, confirming its veracity as a faithful reporter of OX1R expression and utility for future studies assessing the role of OX1R in more complex behaviours.


Subject(s)
Brain Stem/metabolism , Neurons/metabolism , Orexin Receptors/biosynthesis , Prosencephalon/cytology , Prosencephalon/metabolism , Animals , Brain Stem/cytology , Green Fluorescent Proteins , Immunohistochemistry , Male , Mice , Mice, Transgenic , Orexin Receptors/analysis , Polymerase Chain Reaction
10.
Neuropeptides ; 50: 9-16, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25817882

ABSTRACT

Orexin 1 receptor signaling plays a significant role in pain as well as learning and memory processes. This study was conducted to assess the changes in orexin 1 receptor expression levels in hippocampus following learning and memory impairment induced by tooth inflammatory pulpal pain. Adult male Wistar rats received intradental injection of 100 µg capsaicin to induce pulpal pain. After recording the pain scores, spatial learning and memory were assessed using Morris Water Maze test. The hippocampal levels of orexin 1 receptor mRNA and protein were determined by semiquantitative reverse transcriptase polymerase chain reaction (RT-PCR) and immunoblotting respectively. The data showed that capsaicin-induced tooth inflammatory pulpal pain was correlated with learning and memory impairment. Intra-hippocampal injection of orexin A inhibited pain-induced learning and memory impairment. However, orexin 1 receptor antagonist, SB-334867, had no effect on learning and memory impairment. Moreover, capsaicin-induced pain significantly decreased hippocampal orexin 1 receptor mRNA and protein levels. Meanwhile, reversed changes took place in the ibuprofen-pretreated group (p < 0.05). It seems that decrease in orexin 1 receptor density and signaling could be involved in tooth pain-induced learning and memory impairment.


Subject(s)
Hippocampus/physiopathology , Learning Disabilities/etiology , Memory Disorders/etiology , Nerve Tissue Proteins/physiology , Orexin Receptors/physiology , Toothache/psychology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzoxazoles/pharmacology , Capsaicin/toxicity , Dental Pulp/drug effects , Hippocampus/metabolism , Ibuprofen/therapeutic use , Incisor , Learning Disabilities/genetics , Learning Disabilities/physiopathology , Male , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/physiopathology , Naphthyridines , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nociception/drug effects , Orexin Receptors/biosynthesis , Orexin Receptors/genetics , Orexins/pharmacology , Pain Measurement , RNA, Messenger/biosynthesis , Random Allocation , Rats , Rats, Wistar , Single-Blind Method , Toothache/drug therapy , Toothache/genetics , Toothache/physiopathology , Urea/analogs & derivatives , Urea/pharmacology
11.
Int J Mol Med ; 35(2): 539-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25515760

ABSTRACT

Orexins are hypothalamic peptides that regulate food intake, wakefulness, the reward system and energy metabolism. Recent studies have demonstrated the ability of orexins to promote a robust apoptosis and subsequent inhibition of cell growth in various types of cancer cells. The present study was conducted to investigate the effects of orexin A on the survival of human gastric cancer cells, SGC­7901, and the possible mechanisms. SGC­7901 cells were exposed to various concentrations of orexin A in vitro in the presence or absence of the orexin receptor 1 (OX1R) antagonist (SB334867), extracellular signal­regulated kinases 1 and 2 (ERK1/2) antagonist (U0126) or a combination of the two antagonists. The amount of cell proliferation, viability and apoptosis, caspase­8 and caspases­9 activities, OX1R protein expression and ERK1/2 protein levels were determined. The expression of OX1R in SGC­7901 cells was observed. Orexin A (10-10 to 10-6 M) stimulated SGC­7901 cell proliferation and viability, reduced the pro­apoptotic activity of caspase­9 and protected the cells from apoptosis in a dose­dependent manner. Additionally, ERK1/2 phosphorylation was stimulated by orexin A (10-10 to 10-6 M). However, the OX1R antagonist SB334867 (10-6 M), ERK1/2 antagonist U0126 (30 µM) or the combination of antagonists blocked the effects of orexin A to a certain extent. These results suggest that stimulation of OX1R induces the growth of SGC­7901 gastric cancer cells through activation of ERK1/2 signaling pathway. These findings add a new dimension to the biological activities of orexin, which may have important implications in health and disease, in particular gastric cancer.


Subject(s)
Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/biosynthesis , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Orexin Receptors/biosynthesis , Stomach Neoplasms/metabolism , Cell Line, Tumor , Humans , Orexins , Stomach Neoplasms/pathology , Up-Regulation/drug effects
12.
Glia ; 62(6): 982-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24639050

ABSTRACT

The mechanisms that control microglial activation are of interest, since neuroinflammation, which involves reactive microglia, may be an additional target in the search for therapeutic strategies to treat neurodegenerative diseases. Neuron-microglia interaction through contact-dependent or independent mechanisms is involved in the regulation of the microglial phenotype in both physiological and pathological conditions. The interaction between CD200, which is mainly present in neurons but also in astrocytes, and CD200R1, which is mainly present in microglia, is one of the mechanisms involved in keeping the microglial proinflammatory phenotype under control in physiological conditions. Alterations in the expression of CD200 and CD200R1 have been described in neurodegenerative diseases, but little is known about the mechanism of regulation of these proteins under physiological or pathological conditions. The aim of this work was to study the modulation of CD200 and CD200R1 expression by peroxisome proliferator-activated receptor gamma (PPAR-γ), a transcription factor involved in the control of the inflammatory response. Mouse primary neuronal and glial cultures and neuron-microglia cocultures were treated with the PPAR-γ endogenous ligand 15-deoxy-Δ(12, 14) -prostaglandin J2 (15d-PGJ2 ) in the presence and absence of lipopolysaccharide plus interferon-γ (LPS/IFN-γ)-induced glial activation. We show that 15d-PGJ2 inhibits the pro-inflammatory response and prevents both CD200R1 downregulation and CD200 upregulation in reactive glial cells. In addition, 15d-PGJ2 abrogates reactive-microglia induced neurotoxicity in neuron-microglia cultures through a CD200-CD200R1 dependent mechanism. These results suggest that PPAR-γ modulates CD200 and CD200R1 gene expression and that CD200-CD200R1 interaction is involved in the anti-inflammatory and neuroprotective action of PPAR-γ agonists.


Subject(s)
Antigens, CD/biosynthesis , Gene Expression Regulation , Neuroglia/metabolism , Orexin Receptors/biosynthesis , PPAR gamma/physiology , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Coculture Techniques , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , PPAR gamma/agonists , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology
13.
Anat Histol Embryol ; 43(1): 42-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23521689

ABSTRACT

Orexins A (ox A) and B are two peptides originally discovered in neurons of rat hypothalamus, and later found in different cellular types of the gastrointestinal and genital tracts. They arise from the proteolytic cleavage of a common precursor molecule, prepro-orexin, and bind to two receptors, namely receptor 1 (ox1r) and receptor 2 for orexins, that show different binding affinity. The central role of the two peptides has been extensively studied, whereas their activity in the periphery is still poorly known. Here, we investigated the presence of ox A and ox1r in the epididymis of a South American camelid species, the alpaca, by immunohistochemistry, and we also assessed the expression of prepro-orexin and ox1r in tissue extracts by Western blotting analysis. Ox A- and ox1r-immunoreactivity was found in the cytoplasm of principal cells of the caput epididymis. A prevalent supranuclear localization of granular-shaped positive material was observed. No positivity was present in the other cytotypes of epididymis. The expression of two peptides with molecular weight corresponding to those of prepro-orexin and ox1r, respectively, was detected in the tissue extracts from the organ.


Subject(s)
Camelids, New World/metabolism , Epididymis/metabolism , Immunohistochemistry/veterinary , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptides/biosynthesis , Orexin Receptors/biosynthesis , Animals , Epididymis/cytology , Male , Orexins , Protein Binding
14.
Psychoneuroendocrinology ; 38(4): 560-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22917622

ABSTRACT

The orexin neuropeptide system regulates wakefulness and contributes to physiological and behavioral stress responses. Moreover, a role for orexins in modulating hypothalamus-pituitary-adrenal (HPA) axis activity has been proposed. Brain penetrating dual orexin receptor (OXR) antagonists such as almorexant decrease vigilance and have emerged as a novel therapeutic class for the treatment of insomnia. Almorexant was used here as a pharmacological tool to examine the role of endogenous orexin signaling in HPA axis endocrine function under natural conditions. After confirming the expression of prepro-orexin and OXR-1 and OXR-2 mRNA in hypothalamus, pituitary and adrenal glands, the effects of systemic almorexant were investigated on peripheral HPA axis hormone release in the rat under baseline, stress and pharmacological challenge conditions. Almorexant did not alter basal or stress-induced corticosterone release despite affecting wake and sleep stages (detected by radiotelemetric electroencephalography/electromyography) during the stress exposure. Moreover, almorexant did not affect the release of adrenocorticotropin (ACTH) and corticosterone at different time points along the diurnal rhythm, nor corticotrophin-releasing hormone (CRH)- and ACTH-stimulated neuroendocrine responses, measured in vivo under stress-free conditions. These results illustrate that dual OXR antagonists, despite modulating stress-induced wakefulness, do not interfere with endocrine HPA axis function in the rat. They converge to suggest that endogenous orexin signaling plays a minor role in stress hormone release under basal conditions and under challenge.


Subject(s)
Acetamides/pharmacology , Adrenal Glands/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Isoquinolines/pharmacology , Neuropeptides/physiology , Orexin Receptor Antagonists , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/blood , Animals , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Corticosterone/blood , Corticotropin-Releasing Hormone/pharmacology , Hypnotics and Sedatives/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Male , Neuropeptides/biosynthesis , Orexin Receptors/biosynthesis , Orexins , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Restraint, Physical/physiology , Sleep Stages/drug effects , Wakefulness/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...