Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 389(4): 309-321, 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37494485

ABSTRACT

BACKGROUND: Narcolepsy type 1 is caused by severe loss or lack of brain orexin neuropeptides. METHODS: We conducted a phase 2, randomized, placebo-controlled trial of TAK-994, an oral orexin receptor 2-selective agonist, in patients with narcolepsy type 1. Patients with confirmed narcolepsy type 1 according to clinical criteria were randomly assigned to receive twice-daily oral TAK-994 (30 mg, 90 mg, or 180 mg) or placebo. The primary end point was the mean change from baseline to week 8 in average sleep latency (the time it takes to fall asleep) on the Maintenance of Wakefulness Test (range, 0 to 40 minutes; normal ability to stay awake, ≥20 minutes). Secondary end points included the change in the Epworth Sleepiness Scale (ESS) score (range, 0 to 24, with higher scores indicating greater daytime sleepiness; normal, <10) and the weekly cataplexy rate. RESULTS: Of the 73 patients, 17 received TAK-994 at a dose of 30 mg twice daily, 20 received 90 mg twice daily, 19 received 180 mg twice daily, and 17 received placebo. The phase 2 trial and an extension trial were terminated early owing to hepatic adverse events. Primary end-point data were available for 41 patients (56%); the main reason for missing data was early trial termination. Least-squares mean changes to week 8 in average sleep latency on the MWT were 23.9 minutes in the 30-mg group, 27.4 minutes in the 90-mg group, 32.6 minutes in the 180-mg group, and -2.5 minutes in the placebo group (difference vs. placebo, 26.4 minutes in the 30-mg group, 29.9 minutes in the 90-mg group, and 35.0 minutes the 180-mg group; P<0.001 for all comparisons). Least-squares mean changes to week 8 in the ESS score were -12.2 in the 30-mg group, -13.5 in the 90-mg group, -15.1 in the 180-mg group, and -2.1 in the placebo group (difference vs. placebo, -10.1 in the 30-mg group, -11.4 in the 90-mg group, and -13.0 in the 180-mg group). Weekly incidences of cataplexy at week 8 were 0.27 in the 30-mg group, 1.14 in the 90-mg group, 0.88 in the 180-mg group, and 5.83 in the placebo group (rate ratio vs. placebo, 0.05 in the 30-mg group, 0.20 in the 90-mg group, and 0.15 in the 180-mg group). A total of 44 of 56 patients (79%) receiving TAK-994 had adverse events, most commonly urinary urgency or frequency. Clinically important elevations in liver-enzyme levels occurred in 5 patients, and drug-induced liver injury meeting Hy's law criteria occurred in 3 patients. CONCLUSIONS: In a phase 2 trial involving patients with narcolepsy type 1, an orexin receptor 2 agonist resulted in greater improvements on measures of sleepiness and cataplexy than placebo over a period of 8 weeks but was associated with hepatotoxic effects. (Funded by Takeda Development Center Americas; TAK-994-1501 and TAK-994-1504 ClinicalTrials.gov numbers, NCT04096560 and NCT04820842.).


Subject(s)
Narcolepsy , Orexin Receptors , Orexins , Humans , Cataplexy/complications , Cataplexy/drug therapy , Cataplexy/epidemiology , Double-Blind Method , Narcolepsy/drug therapy , Narcolepsy/complications , Narcolepsy/epidemiology , Orexin Receptors/agonists , Orexin Receptors/therapeutic use , Sleepiness/drug effects , Treatment Outcome , Orexins/analysis , Orexins/deficiency , Orexins/pharmacology , Brain Chemistry/drug effects , Administration, Oral , Chemical and Drug Induced Liver Injury/etiology
2.
Nat Methods ; 19(2): 231-241, 2022 02.
Article in English | MEDLINE | ID: mdl-35145320

ABSTRACT

Orexins (also called hypocretins) are hypothalamic neuropeptides that carry out essential functions in the central nervous system; however, little is known about their release and range of action in vivo owing to the limited resolution of current detection technologies. Here we developed a genetically encoded orexin sensor (OxLight1) based on the engineering of circularly permutated green fluorescent protein into the human type-2 orexin receptor. In mice OxLight1 detects optogenetically evoked release of endogenous orexins in vivo with high sensitivity. Photometry recordings of OxLight1 in mice show rapid orexin release associated with spontaneous running behavior, acute stress and sleep-to-wake transitions in different brain areas. Moreover, two-photon imaging of OxLight1 reveals orexin release in layer 2/3 of the mouse somatosensory cortex during emergence from anesthesia. Thus, OxLight1 enables sensitive and direct optical detection of orexin neuropeptides with high spatiotemporal resolution in living animals.


Subject(s)
Brain/metabolism , Molecular Imaging/methods , Orexin Receptors/genetics , Orexins/analysis , Recombinant Proteins/metabolism , Animals , Behavior, Animal , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Orexin Receptors/metabolism , Orexins/genetics , Orexins/pharmacology , Photons , Recombinant Proteins/genetics , Reproducibility of Results , Sleep/physiology
3.
J Comp Neurol ; 529(5): 929-956, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32678476

ABSTRACT

As stressful environment is a potent modulator of feeding, we seek in the present work to decipher the neuroanatomical basis for an interplay between stress and feeding behaviors. For this, we combined anterograde and retrograde tracing with immunohistochemical approaches to investigate the patterns of projections between the dorsomedial division of the bed nucleus of the stria terminalis (BNST), well connected to the amygdala, and hypothalamic structures such as the paraventricular (PVH) and dorsomedial (DMH), the arcuate (ARH) nuclei and the lateral hypothalamic areas (LHA) known to control feeding and motivated behaviors. We particularly focused our study on afferences to proopiomelanocortin (POMC), agouti-related peptide (AgRP), melanin-concentrating-hormone (MCH) and orexin (ORX) neurons characteristics of the ARH and the LHA, respectively. We found light to intense innervation of all these hypothalamic nuclei. We particularly showed an innervation of POMC, AgRP, MCH and ORX neurons by the dorsomedial and dorsolateral divisions of the BNST. Therefore, these results lay the foundation for a better understanding of the neuroanatomical basis of the stress-related feeding behaviors.


Subject(s)
Amygdala/anatomy & histology , Hypothalamus/anatomy & histology , Mice/anatomy & histology , Neural Pathways/anatomy & histology , Septal Nuclei/anatomy & histology , Agouti-Related Protein/analysis , Animals , Axonal Transport , Feeding Behavior/physiology , Feeding Behavior/psychology , Hypothalamic Hormones/analysis , Luminescent Proteins/analysis , Male , Melanins/analysis , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Neurons/chemistry , Neurons/classification , Neurons/ultrastructure , Orexins/analysis , Phytohemagglutinins/analysis , Pituitary Hormones/analysis , Proprotein Convertases/analysis , Rabies virus , Species Specificity , Tyrosine 3-Monooxygenase/analysis , Red Fluorescent Protein
4.
Rev. neurol. (Ed. impr.) ; 71(12): 460-466, 16 dic., 2020.
Article in Spanish | IBECS | ID: ibc-199340

ABSTRACT

INTRODUCCIÓN: Entre los mediadores químicos que modulan el eje intestino-cerebro debe incluirse el sistema orexinérgico, ya que la orexina A (OXA) hipotalámica interviene en la motilidad y en la secreción gastrointestinal. También está presente en las células enteroendocrinas de la mucosa intestinal y en las neuronas aferentes primarias del plexo mientérico, y puede intervenir en la señalización intestino-cerebro. OBJETIVO: No se conoce con exactitud la fuente ni la señal que originan la liberación de OXA periférica, ni tampoco si actúa en los receptores orexinérgicos de los tejidos periféricos ante demandas fisiológicas o patológicas. Esta revisión intenta analizar estas cuestiones a la luz de nuevos datos que indican que la OXA en el eje intestino-cerebro puede tener funciones más allá de su participación en la homeostasis energética. DESARROLLO: La OXA en el sistema entérico protege de la inflamación sistémica y central, y en el hipotálamo orquesta numerosos efectos periféricos para suprimir la respuesta inflamatoria sistémica. Por ello, podría actuar como sustancia inmunomoduladora en inflamaciones crónicas o en enfermedades autoinmunitarias. La OXA también se relaciona con la respuesta de estrés, regulando las respuestas fisiológicas a estímulos emocionales o estresantes. CONCLUSIONES: Aunque la OXA tiene efectos antiinflamatorios y gastroprotectores de la mucosa intestinal, en procesos de inflamación crónica podría incrementar la respuesta a estímulos estresantes, tanto externos como internos, y exacerbar la inflamación gastrointestinal. Por ello, se han propuesto intervenciones farmacológicas sobre el sistema orexinérgico como tratamiento para enfermedades en las que la hipersensibilidad intestinal coexiste con pérdida de apetito, alteraciones del sueño, estrés y ansiedad


INTRODUCTION. The orexinergic system is one of the chemical mediators that modulate the gut-brain axis, given the involvement of hypothalamic orexin A (OXA) in gastrointestinal motility and secretion, and the presence of OXA in enteroendocrine cells of the intestinal mucosa and in primary afferent neurons of the mesenteric plexus, permitting its participation in gut-brain signaling. AIM. The source of OXA and the signal(s) triggering its peripheral release are not fully understood, and it is not known whether it acts on orexigenic receptors in peripheral tissues to meet physiological or pathological demands. The aim of this review is to address these questions in the light of new data indicating that OXA may have functions in the gut-brain axis that go beyond its participation in energy homeostasis. DEVELOPMENT. OXA in the enteric system protects against systemic and central inflammation, and hypothalamic OXA orchestrates numerous peripheral effects to suppress the systemic inflammatory response. For this reason, OXA may act as an immunomodulator in chronic inflammations or autoimmune diseases. OXA is also involved in the stress response, regulating physiological responses to emotional or stressful stimuli. CONCLUSIONS. OXA exerts anti-inflammatory and gastroprotective effects on the intestinal mucosa; however, it may increase the response to external and/or internal stress in individuals with chronic inflammation, exacerbating the gastrointestinal inflammation. Hence, pharmacologic interventions in the orexinergic system have been proposed to treat diseases in which intestinal hypersensitivity is combined with appetite loss, sleep disturbance, stress, and anxiety


Subject(s)
Humans , Orexins/physiology , Gastrointestinal Tract/metabolism , Immune System/metabolism , Stress, Psychological/metabolism , Neurons/metabolism , Orexins/analysis
5.
Neuropharmacology ; 167: 107993, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32135427

ABSTRACT

Hypocretin/orexin neurons are distributed restrictively in the hypothalamus, a brain region known to orchestrate diverse functions including sleep, reward processing, food intake, thermogenesis, and mood. Since the hypocretins/orexins were discovered more than two decades ago, extensive studies have accumulated concrete evidence showing the pivotal role of hypocretin/orexin in diverse neural modulation. New method of viral-mediated tracing system offers the possibility to map the monosynaptic inputs and detailed anatomical connectivity of Hcrt neurons. With the development of powerful research techniques including optogenetics, fiber-photometry, cell-type/pathway specific manipulation and neuronal activity monitoring, as well as single-cell RNA sequencing, the details of how hypocretinergic system execute functional modulation of various behaviors are coming to light. In this review, we focus on the function of neural pathways from hypocretin neurons to target brain regions. Anatomical and functional inputs to hypocretin neurons are also discussed. We further briefly summarize the development of pharmaceutical compounds targeting hypocretin signaling. This article is part of the special issue on Neuropeptides.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Nerve Net/chemistry , Nerve Net/metabolism , Orexins/metabolism , Animals , Behavior, Addictive/genetics , Behavior, Addictive/metabolism , Eating/physiology , Humans , Optogenetics/methods , Optogenetics/trends , Orexins/analysis , Orexins/genetics , Sleep/physiology
6.
J Comp Neurol ; 526(18): 2937-2954, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30019757

ABSTRACT

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.


Subject(s)
Hypothalamus/cytology , Neural Pathways/cytology , Neurons/cytology , Animals , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Pathways/metabolism , Neurons/metabolism , Orexins/analysis , Orexins/biosynthesis
7.
Eur Rev Med Pharmacol Sci ; 22(5): 1499-1505, 2018 03.
Article in English | MEDLINE | ID: mdl-29565513

ABSTRACT

OBJECTIVE: To explore the effect of parecoxib on cerebral infarction in rats and the regulatory mechanism on hypothalamus orexin neurons (orexin) and protein expression. MATERIALS AND METHODS: 60 SD male rats were randomly divided into sham operation group, model group and treatment group (20 rats in each group). Cerebral infarction model was established by modified Longa method. Rats in the treatment group were given parecoxib (2.5 mg kg-1) in tail by intravenous injection, while both the sham operation group and the model group were given the equal volume of sterile PBS solution in the tail vein. Continuous intervention of 72h was carried out in the three groups. Immunofluorescence staining and Western blot were used to detect the expression of orexin neurons and orexin protein in the hypothalamus of rats, respectively. RESULTS: Immunofluorescence staining showed that the number of orexin positive cells in the model group was significantly less than that in the sham-operated group (p < 0.01). After treatment intervention, the number of orexin positive cells in the hypothalamus was significantly increased compared to that in model group (p < 0.01). Western blot analysis showed that compared with sham operation group, the expression of orexin in the hypothalamus of model group was significantly decreased (p < 0.01), whereas the expression of orexin protein was significantly elevated after parecoxib intervention (p < 0.01). CONCLUSIONS: Parecoxib plays a therapeutic effect on cerebral infarction by up-regulating the orexin neuron.


Subject(s)
Cerebral Infarction/drug therapy , Hypothalamus/drug effects , Isoxazoles/pharmacology , Orexins/analysis , Animals , Hypothalamus/chemistry , Isoxazoles/therapeutic use , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley
9.
Sleep Med ; 33: 57-60, 2017 05.
Article in English | MEDLINE | ID: mdl-28449906

ABSTRACT

OBJECTIVES: The temporal association between sudden infant death syndrome (SIDS) and sleep suggests that the arousability from sleep provides a protective mechanism for survival. Recently, the hypocretin system, which promotes wakefulness, has been implicated in SIDS, since it has been reported that SIDS victims have fewer hypocretin neurons than infants who have died from other causes. To understand the role of hypocretin in SIDS, it is essential to better understand how this system matures. The present study compared cerebrospinal fluid (CSF) hypocretin in children aged 2-6 months, which is the age of peak incidence for SIDS, to both younger and older children. METHOD: Hypocretin levels were measured in CSF samples from 101 children who underwent a clinically relevant lumbar puncture. Children were separated into five age groups: 0-2 months, 2-6 months, 1-5 years, 5-10 years, and 10-18 years. RESULTS: Hypocretin levels were not significantly different between 1-5 years, 5-10 years, and 10-18 years. Therefore, these three groups were pooled into a single one (1-18 years) for further analysis. Between the 0-2 month, 2-6 month, and 1-18 year groups, a significant difference in CSF hypocretin levels existed (p = 0.001). Simple comparisons showed that CSF hypocretin levels in the 2-6 month age group were significantly lower than hypocretin levels in both the 0-2 month and 1-18 year group (p < 0.001 and p = 0.008, respectively), but not significantly between 0-2 month and 1-18 year children. CONCLUSIONS: The CSF hypocretin levels were lower at the age of peak incidence for SIDS. This could underlie an increased vulnerability to SIDS at this specific age.


Subject(s)
Orexins/analysis , Sleep/physiology , Sudden Infant Death/cerebrospinal fluid , Wakefulness/physiology , Adolescent , Child , Child, Preschool , Female , Humans , Incidence , Infant , Infant, Newborn , Male , Spinal Puncture/methods , Sudden Infant Death/epidemiology , Sudden Infant Death/etiology
10.
J Chem Neuroanat ; 82: 12-28, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28416460

ABSTRACT

The nuclear organization of the cholinergic, catecholaminergic, serotonergic and orexinergic neurons in the brains of two species of carnivore, the banded mongoose (Mungos mungo) and domestic ferret (Mustela putorius furo), is presented. The banded mongoose belongs to the feliform suborder and the domestic ferret to the caniform suborder, having last shared a common ancestor approximately 53 million years ago; however, they have a very similar overall morphology and life history, presenting an interesting opportunity to examine the extent of evolutionary plasticity in these systems. The brains of the two carnivore species were coronally sectioned and immunohistochemically stained with antibodies against choline acetyltransferase, tyrosine hydroxylase, serotonin and orexin-A. The overall organization and complement of the nuclei of these systems was identical between the two species, although minor differences were noted. Moreover, this overall organization is identical to other studies undertaken in the domestic cat and dog. While for the most part the nuclei forming these systems are similar to those observed in other mammals, two species differences, which appear to be carnivore-specific, were noted. First, cholinergic neurons were observed in the lateral septal nucleus of both species, an apparently carnivore specific feature not recorded previously in other mammals. Second, the serotonergic neurons of the peripheral division of the dorsal raphe complex exhibited a significant caudad expansion, intermingling with the cholinergic and catecholaminergic nuclei of the pons, a carnivore specific feature. These carnivore specific features likely have functional consequences related to coping with stress and the expression of sleep.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Catecholamines/metabolism , Cholinergic Neurons/metabolism , Orexins/metabolism , Serotonergic Neurons/metabolism , Animals , Catecholamines/analysis , Cholinergic Neurons/chemistry , Ferrets , Herpestidae , Male , Neurons/chemistry , Neurons/metabolism , Orexins/analysis , Serotonergic Neurons/chemistry , Species Specificity
11.
J Histochem Cytochem ; 65(3): 153-171, 2017 03.
Article in English | MEDLINE | ID: mdl-28080173

ABSTRACT

Storage of tissue sections for long periods allows multiple samples, acquired over months or years, to be processed together, in the same reagents, for quantitative histochemical studies. Protocols for freezer storage of free-floating frozen sections using sucrose with different additives have been reported and assert that storage has no effect on histochemistry, but no quantitative support has been provided. The present study analyzed the efficacy of long-term storage of brain tissue sections at -80C in buffered 15% glycerol. To determine whether histochemical reactivity is affected, we analyzed 11 datasets from 80 monkey brains that had sections stored for up to 10 years. For processing, sections from multiple cases were removed from storage, thawed, and batch-processed at the same time for different histochemical measures, including IHC for neuronal nuclear antigen, parvalbumin, orexin-A, doublecortin, bromodeoxyuridine, the pro-form of brain-derived neurotrophic factor, and damaged myelin basic protein as well as a histochemical assay for hyaluronic acid. Results were quantified using stereology, optical densitometry, fluorescence intensity, or percent area stained. Multiple regression analyses controlling for age and sex demonstrated the general stability of these antigens for up to a decade when stored in 15% glycerol at -80C.


Subject(s)
Brain Chemistry , Frozen Sections/methods , Animals , Antigens, Nuclear/analysis , Brain-Derived Neurotrophic Factor/analysis , Bromodeoxyuridine/analysis , Cell Count , Cryopreservation/methods , Doublecortin Domain Proteins , Female , Hyaluronic Acid/analysis , Immunohistochemistry/methods , Macaca mulatta , Male , Microtubule-Associated Proteins/analysis , Myelin Basic Protein/analysis , Nerve Tissue Proteins/analysis , Neuropeptides/analysis , Orexins/analysis , Parvalbumins/analysis
12.
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
13.
Neurosci Lett ; 627: 155-9, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27264485

ABSTRACT

Hypocretin-1 (HC, orexin-A) is a neuropeptide involved in regulating physiological functions of sleep, appetite and arousal, and it has been shown that intranasal (IN) administration can target HC to the brain. Recent clinical studies have shown that IN HC has functional effects in human clinical trials. In this study, we use rats to determine whether IN HC has an immediate effect on food consumption and locomotor activity, whether distribution in the brain after IN delivery is dose-dependent, and whether MAPK and PDK1 are affected after IN delivery. Food intake and wheel-running activity were quantified for 24h after IN delivery. Biodistribution was determined 30min after IN delivery of both a high and low dose of 125I-radiolabelled HC throughout the brain and other bodily tissues, while Western blots were used to quantify changes in cell signaling pathways (MAPK and PDK1) in the brain. Intranasal HC significantly increased food intake and wheel activity within 4h after delivery, but balanced out over the course of 24h. The distribution studies showed dose-dependent delivery in the CNS and peripheral tissues, while PDK1 was significantly increased in the brain 30min after IN delivery of HC. This study adds to the growing body of evidence that IN administration of HC is a promising strategy for treatment of HC related behaviors.


Subject(s)
Eating/drug effects , Motor Activity/drug effects , Orexins/administration & dosage , Administration, Intranasal , Animals , Brain Chemistry , Drinking/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Orexins/analysis , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord/chemistry
14.
Arch Pediatr ; 23(6): 603-6, 2016 Jun.
Article in French | MEDLINE | ID: mdl-27133373

ABSTRACT

Narcolepsy is a disabling disorder, characterized by excessive daytime sleepiness, irresistible sleep attacks, and partial or complete cataplexy. Many cases of obesity and precocious puberty have been reported in narcoleptic children, suggesting that the deficiency of hypocretin in narcolepsy could also be implicated in appetite stimulation. We report the observations of two young girls, who were referred for obesity and who developed narcolepsy accompanied by an abrupt weight gain. In both cases, specific drugs promoted wakefulness and overweight stabilization. Narcolepsy has to be suspected in sleepy obese children and not misdiagnosed as obstructive apnea. A nocturnal polysomnography with multiple sleep latency tests should be performed to confirm the diagnosis and begin specific treatment that is effective for sleep disorders and weight gain.


Subject(s)
Narcolepsy/complications , Narcolepsy/diagnosis , Pediatric Obesity/complications , Adolescent , Child , Female , Humans , Orexins/analysis , Polysomnography
15.
Sleep Med ; 19: 17-22, 2016 03.
Article in English | MEDLINE | ID: mdl-27198942

ABSTRACT

OBJECTIVE: To follow and analyze the clinical course and quality of life of Pandemrix H1N1-vaccine-related narcolepsy (pNT1). METHODS: Twenty-six drug-naïve confirmed pNT1 subjects completed Epworth Sleepiness Scale (ESS), Ullanlinna Narcolepsy Scale (UNS), Swiss Narcolepsy Scale (SNS), Rimon's Brief Depression scale (RDS), and WHO-5 Well-being index questionnaires near the disease onset and in a follow-up a minimum of two years later. The number of cataplexies and body mass index (BMI) were recorded. The effects of hypocretin-1 levels and sleep recording results were analyzed. The findings at the follow-up visit were compared with 25 non-vaccine-related type 1 narcolepsy (NT1) subjects. RESULTS: In pNT1, RDS score decreased significantly (mean 10.2, SD 4.7 vs mean 6.7, SD 4.5, p = 0.003). Median of BMI increased from 20.8 kg m(-2) to 23.4 kg m(-2), p <0.001. There were no significant differences in other sleep scores. However, deviation and range in questionnaire scores at the follow-up were wide. Subjects with very low or undetectable hypocretin-1 levels had worse scores in UNS (mean 26.4, SD 6.95 vs mean 19.1, SD 3.83, p = 0.006) and ESS (mean 17.9, SD = 4.29 vs mean 14.1, SD = 3.70, p = 0.047) than those with hypocretin-1 levels of 20-110 pg/mL. Most disabling symptoms were excessive daytime sleepiness and disturbed sleep. There were no significant differences between the scores in pNT1 and NT1. CONCLUSIONS: Clinical course of pNT1 is heterogeneous but the evolution of pNT1 seems similar to NT1. Lower hypocretin levels in pNT1 are associated with a more severe phenotype.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/adverse effects , Narcolepsy/diagnosis , Surveys and Questionnaires , Adolescent , Female , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Male , Narcolepsy/cerebrospinal fluid , Orexins/analysis , Orexins/cerebrospinal fluid , Polysomnography/methods
16.
Bioorg Med Chem Lett ; 25(15): 2875-87, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26045032

ABSTRACT

Specific neurons in the lateral hypothalamus produce the orexin neuropeptides (orexin-A and orexin-B). The orexin-peptides are transported to areas of the brain regulating sleep-wake cycles, controlling food intake or modulating emotional states such as panic or anxiety. The orexin system, consisting of the two orexin-neuropeptides and two G-protein-coupled receptors (the orexin-1 and the orexin-2 receptor) is as well involved in reward and addictive behaviors. The review reflects on the most recent activities in the field of orexin research.


Subject(s)
Brain/physiology , Orexin Receptors/metabolism , Orexins/metabolism , Sleep Wake Disorders/metabolism , Sleep , Animals , Anxiety/drug therapy , Anxiety/metabolism , Brain/drug effects , Drug Discovery , Humans , Orexin Receptors/analysis , Orexins/analysis , Sleep/drug effects , Sleep Wake Disorders/drug therapy , Wakefulness/drug effects
17.
Alcohol Clin Exp Res ; 39(1): 21-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25623402

ABSTRACT

BACKGROUND: Orexin (OX) neurons originating in the lateral hypothalamus (LH) are ideally positioned to modulate reward processing as they form connections with several key brain regions known to be involved in the reward pathway. Consistent with these findings, a growing number of studies have implicated the OX system in modulating the rewarding properties of several drugs of abuse, including ethanol (EtOH). However, the role of the OX system in excessive binge-like EtOH intake remains relatively unexplored. Here, we assessed changes in OX immunoreactivity (IR) in the hypothalamus following repeated cycles of binge-like EtOH drinking and assessed the participation of the OX-1 receptor (OX1R) in binge-like EtOH consumption. METHODS: The drinking-in-the-dark (DID) paradigm was used to model binge-like EtOH drinking in male C57BL/6J mice. In the first experiment, mice experienced 1 or 3 cycles of binge-like EtOH or sucrose drinking with DID procedures to assess changes in OX IR in distinct subregions of the hypothalamus. Subsequent experiments examined binge-like EtOH and saccharin drinking following peripheral injections of 0.0, 5.0, or 10.0 mg/kg SB-334867 (SB), a selective OX1R antagonist. Finally, mice were given peripheral injections of SB and open-field locomotor activity was measured. RESULTS: Relative to water drinking controls, binge-like consumption of EtOH and sucrose resulted in a marked reduction in OX IR in the LH. Inhibition of the OX1R via SB blunted EtOH and saccharin drinking, but did not alter open-field locomotor activity. CONCLUSIONS: Our observed reduction in OX IR in the LH indicates that the OX system in engaged during binge-like consumption of EtOH and sucrose. The observation that inhibition of the OX1R signaling blunted binge-like EtOH, and saccharin drinking suggests that reward-related OX circuits originating in the LH participate in the consumption of salient reinforcers regardless of calories.


Subject(s)
Binge Drinking/drug therapy , Binge Drinking/metabolism , Hypothalamic Area, Lateral/drug effects , Orexin Receptors/metabolism , Orexins/metabolism , Reinforcement, Psychology , Saccharin/pharmacology , Sucrose/pharmacology , Animals , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Dose-Response Relationship, Drug , Hypothalamic Area, Lateral/metabolism , Male , Mice , Motor Activity/drug effects , Naphthyridines , Orexins/analysis , Orexins/immunology , Sucrose/antagonists & inhibitors , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
18.
J Neurotrauma ; 32(5): 289-96, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25242371

ABSTRACT

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.


Subject(s)
Brain Injuries/complications , Sleep Disorders, Circadian Rhythm/etiology , Sleep Disorders, Circadian Rhythm/physiopathology , Wakefulness/physiology , Animals , Brain Injuries/metabolism , Brain Injuries/physiopathology , Disease Models, Animal , Electroencephalography , Electromyography , Hypothalamus/metabolism , Immunohistochemistry , Male , Orexins/analysis , Orexins/biosynthesis , Rats , Rats, Sprague-Dawley , Sleep Disorders, Circadian Rhythm/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...