Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 222
1.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(2): 254-259, 2024 Apr.
Article Zh | MEDLINE | ID: mdl-38686723

Dopamine,a neurotransmitter ubiquitous in the body fluids,blood,and urine of mammals and humans,is responsible for regulating their functions and metabolism.The dopamine system is involved in the neurobiological mechanisms of narcolepsy in animals and humans.However,researchers have drawn different or even opposite conclusions when measuring the dopamine level in the cerebrospinal fluid of narcolepsy patients.Studies have confirmed that the occurrence of narcolepsy is related to the irreversible loss of orexins.The autoimmune reaction caused by the interactions of environmental factors with genetic factors destroys the hypothalamic orexin neurons and reduces orexin secretion,thereby lowering the level of arousal.We introduce the research progress and current status of dopamine and clinical characterization of narcolepsy by reviewing more than 40 articles published from 1982 to 2023,aiming to provide a reference for studying the relationship between the dopamine level and clinical characterization of narcolepsy and searching for the biomarkers of type 2 narcolepsy.


Dopamine , Narcolepsy , Orexins , Humans , Narcolepsy/metabolism , Narcolepsy/diagnosis , Dopamine/metabolism , Orexins/metabolism , Orexins/cerebrospinal fluid , Animals , Neuropeptides/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
2.
Neurology ; 102(10): e209326, 2024 May.
Article En | MEDLINE | ID: mdl-38669634

BACKGROUND AND OBJECTIVES: Narcolepsy type 1 (NT1) is due to the loss of hypothalamic neurons that produce orexin (ORX), by a suspected immune-mediated process. Rare postmortem studies are available and failed to detect any inflammation in the hypothalamic region, but these brains were collected years after the first symptoms. In vivo studies close to disease onset are lacking. We aimed to explore microglia density in the hypothalamus and thalamus in NT1 compared with controls using [18F]DPA-714 PET and to study in NT1 the relationships between microglia density in the hypothalamus and in other regions of interest (ROIs) with disease duration, severity, and ORX levels. METHODS: Patients with NT1 and controls underwent a standardized clinical evaluation and [18F]DPA-714 PET imaging using a radiolabeled ligand specific to the 18 kDa translocator protein (TSPO). TSPO genotyping determined receptor affinity. Images were processed on peripheral module interface using standard uptake value (SUV) on ROIs: hypothalamus, thalamus, frontal area, cerebellum, and the whole brain. SUV ratios (SUVr) were calculated by normalizing SUV with cerebellum uptake. RESULTS: A total of 41 patients with NT1 (21 adults, 20 children, 10 with recent disease onset <1 year) and 35 controls were included, with no significant difference between groups for [18F]DPA-714 binding (SUV/SUVr) in the hypothalamus and thalamus. Unexpectedly, significantly lower SUVr in the whole brain was found in NT1 compared with controls (0.97 ± 0.06 vs 1.08 ± 0.22, p = 0.04). The same finding between NT1 and controls in the whole brain was observed in those with high or mixed TSPO affinity (p = 0.03 and p = 0.04). Similar trend was observed in the frontal area in NT1 (0.96 ± 0.09 vs 1.09 ± 0.25, p = 0.05). In NT1, no association was found between SUVr in different ROIs and age, disease duration, severity, or ORX levels. DISCUSSION: We found no evidence of in vivo increased microglia density in NT1 compared with controls, even close to disease onset, and even unexpectedly a decrease in the whole brain of these patients. These findings do not support the presence of neuroinflammation in the destruction process of ORX neurons. TRIAL REGISTRATION INFORMATION: ClinicalTrials.org NCT03754348.


Microglia , Narcolepsy , Orexins , Positron-Emission Tomography , Humans , Male , Female , Microglia/metabolism , Narcolepsy/metabolism , Narcolepsy/genetics , Narcolepsy/diagnostic imaging , Orexins/metabolism , Adult , Young Adult , Thalamus/metabolism , Thalamus/diagnostic imaging , Pyrazoles , Hypothalamus/metabolism , Hypothalamus/diagnostic imaging , Hypothalamus/pathology , Severity of Illness Index , Middle Aged , Pyrimidines , Adolescent , Receptors, GABA/metabolism , Receptors, GABA/genetics
3.
Amino Acids ; 55(10): 1247-1259, 2023 Oct.
Article En | MEDLINE | ID: mdl-37689600

Narcolepsy is a chronic and underrecognized sleep disorder characterized by excessive daytime sleepiness and cataplexy. Furthermore, narcolepsy type 1 (NT1) has serious negative impacts on an individual's health, society, and the economy. Currently, many sleep centers lack the means to measure orexin levels in the cerebrospinal fluid. We aimed to analyze the characteristics of metabolite changes in patients with NT1, measured by ultra-performance liquid chromatography-tandem mass spectrometry. A principal component analysis (PCA), an orthogonal partial least square discriminant analysis (OPLS-DA), t tests, and volcano plots were used to construct a model of abnormal metabolic pathways in narcolepsy. We identified molecular changes in serum specimens from narcolepsy patients and compared them with control groups, including dehydroepiandrosterone, epinephrine, N-methyl-D-aspartic acid, and other metabolites, based on an OPLS-loading plot analysis. Nine metabolites yielded an area under the receiver operating curve > 0.75. Meanwhile, seven abnormal metabolic pathways were correlated with differential metabolites, such as metabolic pathways; neuroactive ligand‒receptor interaction; and glycine, serine, and threonine metabolism. To our knowledge, this is the first study to reveal the characteristic metabolite changes in sera from NT1 patients for the selection of potential blood biomarkers and the elucidation of NT1 pathogenesis.


Narcolepsy , Tandem Mass Spectrometry , Humans , Narcolepsy/metabolism , Metabolomics , Chromatography, Liquid , Biomarkers
4.
Sleep ; 46(9)2023 09 08.
Article En | MEDLINE | ID: mdl-37155728

Long-term use of sodium oxybate (SXB), (also called gamma-hydroxybutyrate [GHB]) attenuates the cataplexy and sleepiness of human narcolepsy. We had previously found that chronic opiate usage in humans and long-term opiate administration to mice significantly increased the number of detected hypocretin/orexin (Hcrt) neurons, decreased their size, and increased Hcrt level in the hypothalamus. We also found that opiates significantly decreased cataplexy in human narcoleptics as well as in narcoleptic mice and that cessation of locus coeruleus neuronal activity preceded and was tightly linked to cataplectic attacks in narcoleptic dogs. We tested the hypothesis that SXB produces changes similar to opiates and now report that chronic SXB administration significantly increased the size of Hcrt neurons, the reverse of what we had seen with opiates in humans and mice. Levels of Hcrt in the hypothalamus were nonsignificantly lower, in contrast to the significant increase in hypothalamic Hcrt level after opiates. SXB decreased tyrosine hydroxylase levels in the locus coeruleus, the major descending projection of the hypocretin system, also the reverse of what we saw with opioids. Therefore despite some similar effects on narcoleptic symptomatology, SXB does not produce anatomical changes similar to those elicited by opiates. Analysis of changes in other links in the cataplexy pathway might further illuminate SXB's mechanism of action on narcolepsy.


Cataplexy , Narcolepsy , Opiate Alkaloids , Sodium Oxybate , Humans , Mice , Animals , Dogs , Orexins/metabolism , Sodium Oxybate/pharmacology , Cataplexy/drug therapy , Cataplexy/metabolism , Locus Coeruleus/metabolism , Narcolepsy/drug therapy , Narcolepsy/metabolism , Neurons/metabolism , Opiate Alkaloids/metabolism
5.
Peptides ; 165: 171007, 2023 07.
Article En | MEDLINE | ID: mdl-37030519

Orexin (also known as hypocretin) is a neuropeptide exclusively synthesized in the neurons of the lateral hypothalamus (LH). Initially orexin was thought to be involved in the regulation of feeding behavior. However, it is now known to also be a critical regulator of sleep/wakefulness, especially the maintenance of wakefulness. Although the somas of orexin neurons are exclusively located in the LH, these neurons send axons throughout the brain and spinal cord. Orexin neurons integrate inputs from various brain regions and project to neurons that are involved in the regulation of sleep/wakefulness. Orexin knockout mice have a fragmentation of sleep/wakefulness and cataplexy-like behavior arrest, which is similar to the sleep disorder narcolepsy. Recent progress with manipulation of neural activity of targeted neurons, using experimental tools such as optogenetics and chemogenetics, has emphasized the role of orexin neuron activity on the regulation of sleep/wakefulness. Recording of orexin neuron activity in vivo using electrophysiological and gene-encoded calcium indicator proteins revealed that these cells have specific activity patterns across sleep/wakefulness state changes. Here, we also discuss not only the role of the orexin peptide, but also the role of other co-transmitters that are synthesized and released from orexin neurons and involved in sleep/wakefulness regulation.


Narcolepsy , Neuropeptides , Mice , Animals , Orexins/metabolism , Wakefulness/physiology , Sleep/physiology , Neuropeptides/metabolism , Narcolepsy/metabolism , Neurons/metabolism , Mice, Knockout , Orexin Receptors/metabolism
6.
Sleep ; 45(12)2022 Dec 12.
Article En | MEDLINE | ID: mdl-35640640

Three papers currently published in SLEEP using two different mouse models of narcolepsy, including either Hcrt-tTa;TetO diptheria toxin-A (DTA) or Hypocretin knock-out (Hcrt-KO) mice, suggest important gender differences in narcolepsy expression. Specifically, these recent data corroborate previous findings in mice demonstrating that females show more cataplexy events and more total cataplexy expression than males. Moreover, in the neurotoxic DTA mouse model, females show earlier onset of cataplexy expression than males during active Hcrt cell loss. Finally, females show a doubling of cataplexy during estrous compared to other phases of the estrous cycle. These findings are reviewed in the broader context of prior published literature, including reported gender differences in Hcrt expression and hormonal influences on sleep and wakefulness. Although similar findings have not been reported in humans, a systematic evaluation of gender differences in human narcolepsy has yet to be performed. Taken together, these animal data suggest that more research exploring gender differences in human narcolepsy is warranted.


Cataplexy , Narcolepsy , Neuropeptides , Male , Female , Animals , Mice , Humans , Narcolepsy/genetics , Narcolepsy/metabolism , Cataplexy/genetics , Orexins/genetics , Orexins/metabolism , Wakefulness , Sleep , Mice, Knockout , Disease Models, Animal
7.
Mol Brain ; 15(1): 21, 2022 03 04.
Article En | MEDLINE | ID: mdl-35246205

It is unclear why orexin-deficient animals, but not wild-type mice, show cataplexy. The current hypothesis predicts simultaneous excitation of cataplexy-inhibiting orexin neurons and cataplexy-inducing amygdala neurons. To test this hypothesis, we measured the activity of putative orexin neurons in orexin-knockout mice during cataplexy episodes using fiber photometry. We created two animal models of orexin-knockout mice with a GCaMP6 fluorescent indicator expressed in putative orexin neurons. We first prepared orexin-knockout mice crossed with transgenic mice carrying a tetracycline-controlled transactivator transgene under the control of the orexin promoter. TetO-GCaMP6 was then introduced into mice via an adeno-associated virus injection or natural crossing. The resulting two models showed restricted expression of GCaMP6 in the hypothalamus, where orexin neurons should be located, and showed excitation to an intruder stress that was similar to that observed in orexin-intact mice in our previous study. The activity of these putative orexin neurons increased immediately before the onset of cataplexy-like behavior but decreased (approximately - 20% of the baseline) during the cataplexy-like episode. We propose that the activity of orexin neurons during cataplexy is moderately inhibited by an unknown mechanism. The absence of cataplexy in wild-type mice may be explained by basal or residual activity-induced orexin release, and emotional stimulus-induced counter activation of orexin neurons may not be necessary. This study will serve as a basis for better treatment of cataplexy in narcolepsy patients.


Cataplexy , Narcolepsy , Animals , Cataplexy/metabolism , Cataplexy/therapy , Humans , Mice , Mice, Knockout , Mice, Transgenic , Narcolepsy/metabolism , Narcolepsy/therapy , Neurons/metabolism , Orexins/metabolism
8.
Brain Pathol ; 32(2): e13027, 2022 03.
Article En | MEDLINE | ID: mdl-34672414

An increased number of histaminergic neurons, identified by labeling histidine-decarboxylase (HDC) its synthesis enzyme, was unexpectedly found in patients with narcolepsy type 1 (NT1). In quest for enlightenment, we evaluate whether an increase in HDC cell number and expression level would be detected in mouse models of the disease, in order to provide proof of concepts reveling possible mechanisms of compensation for the loss of orexin neurons, and/or of induced expression as a consequence of local neuroinflammation, a state that likely accompanies NT1. To further explore the compensatory hypothesis, we also study the noradrenergic wake-promoting system. Immunohistochemistry for HDC, orexin, and melanin-concentrating hormone (MCH) was used to count neurons. Quantitative-PCR of HDC, orexin, MCH, and tyrosine-hydroxylase was performed to evaluate levels of mRNA expression in the hypothalamus or the dorsal pons. Both quantifications were achieved in genetic and neuroinflammatory models of narcolepsy with major orexin impairment, namely the orexin-deficient (Orex-KO) and orexin-hemagglutinin (Orex-HA) mice respectively. The number of HDC neurons and mRNA expression level were unchanged in Orex-KO mice compared to controls. Similarly, we found no change in tyrosine-hydroxylase mRNA expression in the dorsal pons between groups. Further, despite the presence of protracted local neuroinflammation as witnessed by the presence of reactive microglia, we found no change in the number of neurons nor the expression of HDC in Orex-HA mice compared to controls. Importantly, no correlation was found in all conditions between HDC and orexin. Our findings indicate that, in mice, the expression of histamine and noradrenalin, two wake-promoting systems, are not modulated by orexin level whether the lack of orexin is constitutive or induced at adult age, showing thus no compensation. They also show no recruitment of histamine by local neuroinflammation. Further studies will be needed to further define the role of histamine in the pathophysiology of NT1.


Histamine , Narcolepsy , Animals , Histamine/metabolism , Histidine Decarboxylase/genetics , Humans , Mice , Mixed Function Oxygenases , Narcolepsy/genetics , Narcolepsy/metabolism , Orexins/metabolism , RNA, Messenger
9.
Eur J Nucl Med Mol Imaging ; 49(4): 1127-1135, 2022 03.
Article En | MEDLINE | ID: mdl-34651222

PURPOSE: Histamine H3 receptor antagonists and inverse agonists have been extensively developed to treat sleep-wake, neurocognitive, and allied disorders. However, potential adverse effects, including insomnia, hampered the clinical use of these drugs, possibly due to their persistent interaction with the target molecules. The purpose of the present study was to estimate the pharmacokinetics and pharmacodynamics of enerisant, a novel antagonist and inverse agonist for histamine H3 receptors. METHODS: To measure the histamine H3 receptor occupancy by enerisant, positron emission tomography studies using [11C]TASP457, a specific radioligand for histamine H3 receptors, were performed in 12 healthy men at baseline and at 2 h after oral administration of enerisant hydrochloride. For three of these subjects, two additional scans were performed at 6 and 26 h after the administration. Relationships between the receptor occupancy by enerisant and its dose and plasma concentrations were then analyzed. RESULTS: Administration of enerisant hydrochloride decreased the radioligand binding in a dose-dependent manner. The estimated receptor occupancy values at 2 h varied as a function of its dose or plasma concentration. The time course of the occupancy showed persistently high levels (> 85%) in the two subjects with higher doses (25 and 12.5 mg). The occupancy was also initially high at 2 h and 6 h with the lower dose of 5 mg, but it decreased to 69.7% at 26 h. CONCLUSION: The target engagement of enerisant was demonstrated in the brains of living human subjects. The occupancy of histamine H3 receptors by enerisant at 2 h can be predicted by applying the plasma concentration of enerisant to Hill's plot. The preliminary time-course investigation showed persistently high brain occupancy with high doses of enerisant despite the decreasing plasma concentration of the drug. Five milligrams or less dose would be appropriate for the treatment for narcolepsy with initially high occupancy allowing for effective treatment of narcolepsy, and then the occupancy level would be expected to decrease to a level to avoid this drug's unwanted side effect of insomnia at night, although further research is warranted to confirm the statement since the expected decrease is based on the finding in one subject. TRIAL REGISTRATION: This study was retrospectively registered with ClinicalTrials.gov (NCT04631276) on November 17, 2020.


Narcolepsy , Neuroprotective Agents , Receptors, Histamine H3 , Sleep Initiation and Maintenance Disorders , Brain/diagnostic imaging , Brain/metabolism , Histamine/metabolism , Humans , Ligands , Male , Narcolepsy/metabolism , Niacinamide , Positron-Emission Tomography/methods , Pyridines , Quinolones , Receptors, Histamine H3/metabolism , Sleep Initiation and Maintenance Disorders/metabolism
10.
Front Neurol Neurosci ; 45: 103-116, 2021.
Article En | MEDLINE | ID: mdl-34052809

Since its description in the 19th century, narcolepsy type 1 (NT1) has been considered as a model sleep disorder, and after the discovery of rapid eye movement (REM) sleep onset in the disorder, a gateway to understanding REM sleep. The discovery that NT1 is caused by hypocretin/orexin deficiency, together with neurochemical studies of this system, has helped to establish how this neuropeptide regulates the organization of sleep and wake in humans. Current analyses suggest that the main functions of the hypocretin/orexin system are (1) maintenance of wakefulness in the face of moderate sleep deprivation; (2) passive wake promotion, especially in the evening, driven by the circadian clock; (3) inhibition of REM sleep, with possible differential modulating effects on various subcomponents of the sleep-stage, explaining REM sleep dissociation events in NT1. Narcolepsy is also associated with an inability to consolidate sleep, a more complex phenotype that may result from secondary changes or be central to the role of hypocretin in coordinating the activity of other sleep- and wake-promoting systems. Novel technologies, such as the use of deep learning analysis of electroencephalographic signals, is revealing a complex pattern of sleep abnormalities in human narcolepsy that can be used diagnostically. The availability of novel devices measuring sleep 24 h per day also holds promise to provide new insights into how brain electrical activity and muscle tone are regulated by hypocretin.


Narcolepsy/metabolism , Narcolepsy/physiopathology , Orexins/deficiency , Sleep Stages/physiology , Humans
11.
Front Neurol Neurosci ; 45: 139-149, 2021.
Article En | MEDLINE | ID: mdl-34052817

Advances in translational research provide key opportunities to explore the physiological and pathological effects of sleep in different neurodegenerative diseases. Recent findings suggest that sleep-wakefulness dysfunctions may predispose to neurodegenerative disorders such as Alzheimer's disease (AD), and vice versa. New theories on the link between sleep and ß-amyloid and tau secretion, accumulation and clearance, and its interaction with hypocretins/orexins (key neuropeptides regulating wakefulness) suggest mechanistic ways to better understand the impact of sleep alterations in the pathogenesis of AD. Further studies should validate whether changes in circadian rhythm and sleep-wakefulness patterns could be used for early AD diagnosis and as prognostic markers for cognitive decline. Longitudinal studies are needed, not only to validate these biomarker interactions and to determine the cause-effect relationship and the role of sleep-wakefulness behavior in the regulation of amyloid plaque and neurofibrillary tangle formation, but also to identify the best sleep therapies and related preventive strategies for AD.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Glymphatic System/metabolism , Narcolepsy/metabolism , Orexins/metabolism , Sleep Wake Disorders/metabolism , tau Proteins/metabolism , Animals , Humans , Orexins/deficiency
13.
Biochem Pharmacol ; 191: 114386, 2021 09.
Article En | MEDLINE | ID: mdl-33359009

The circadian wake drive is a mathematic representation of the observed increased propensity to stay awake late in the day, peaking in the hours just before anticipated bed time. It has been called the "forbidden zone" due to the difficulty in initiating sleep during this time and is responsible for the problems initiating sleep when traveling eastward, for maintaining daytime sleep in shift workers, and for initiating sleep in some individuals with insomnia. Evidence culled from studies in individuals with narcolepsy, who lack production of hypocretin (orexin) neuropeptides, as well as a primate model of human wake consolidation and pharmacologic studies of hypocretin antagonists indicate that hypocretin-1 may be the physiologic instantiation of the circadian wake drive. This review will discuss the evidence in support of this hypothesis.


Circadian Rhythm/physiology , Orexins/metabolism , Sleep/physiology , Wakefulness/physiology , Animals , Homeostasis/physiology , Humans , Narcolepsy/metabolism , Narcolepsy/physiopathology
14.
Bioanalysis ; 12(8): 533-544, 2020 Apr.
Article En | MEDLINE | ID: mdl-32351118

Background: A LC-MS/MS method was validated for the quantification of SUVN-G3031, a novel H3 receptor inverse agonist in clinical development for the treatment of patients with narcolepsy, with and without cataplexy. Methodology: SUVN-G3031 was extracted from plasma following acetonitrile protein precipitation, separated by Ultra HPLC and quantified using positive ESI-MS/MS. Results: The method was linear across the range of 0.1-100 ng ml-1 in plasma. Results for intra and inter-day accuracy were from 99.8 to 104% and precision (%CV) was ≤10.6%. Conclusion: The method was applied to a first-in-human study in healthy volunteers. The method is precise, accurate and highly selective for the quantification of SUVN-G3031 in human plasma.


Morpholines/blood , Narcolepsy/drug therapy , Piperidines/blood , Receptors, Histamine H3/metabolism , Chromatography, High Pressure Liquid , Humans , Molecular Structure , Morpholines/pharmacology , Narcolepsy/metabolism , Piperidines/pharmacology , Tandem Mass Spectrometry
15.
Article En | MEDLINE | ID: mdl-32373062

Narcolepsy type 1 is a neurological sleep-wake disorder caused by the destruction of orexin (hypocretin)-producing neurons. These neurons are particularly located in the lateral hypothalamus and have widespread projections throughout the brain, where they are involved, e.g., in the regulation of the sleep-wake cycle and appetite. Interestingly, a higher prevalence of obesity has been reported in patients with narcolepsy type 1 compared to healthy controls, despite a normal to decreased food intake and comparable physical activity. This suggests the involvement of tissues implicated in total energy expenditure, including skeletal muscle, liver, white adipose tissue (WAT), and brown adipose tissue (BAT). Recent evidence from pre-clinical studies with orexin knock-out mice demonstrates a crucial role for the orexin system in the functionality of brown adipose tissue (BAT), probably through multiple pathways. Since BAT is a highly metabolically active organ that combusts fatty acids and glucose toward heat, thereby contributing to energy metabolism, this raises the question of whether BAT plays a role in the development of obesity and related metabolic diseases in narcolepsy type 1. BAT is densely innervated by the sympathetic nervous system that activates BAT, for instance, following cold exposure. The sympathetic outflow toward BAT is mainly mediated by the dorsomedial, ventromedial, arcuate, and paraventricular nuclei in the hypothalamus. This review focuses on the current knowledge on the role of the orexin system in the control of energy balance, with specific focus on BAT metabolism and adiposity in both preclinical and clinical studies.


Adipose Tissue, Brown/physiology , Adiposity/physiology , Narcolepsy/complications , Narcolepsy/metabolism , Animals , Eating/physiology , Energy Metabolism/physiology , Humans , Orexins/physiology
16.
Cytokine ; 131: 155103, 2020 07.
Article En | MEDLINE | ID: mdl-32315956

BACKGROUND: Narcolepsy is a sleep disorder characterized by a loss of hypocretin neurons in the hypothalamus. Inflammation is proposed as a mechanism for neurodegeneration in narcolepsy. Numerous studies have investigated peripheral cytokine measures in narcoleptic patients, though the results are not conclusive. The current systematic review and meta-analysis aims to address the question of how do serum/plasma cytokine levels change in narcolepsy. METHODS: A systematic search of the literature to July 2019, was conducted to identify studies that measured cytokine levels in patients with narcolepsy, compared with those in controls without narcolepsy. RESULTS: Twelve studies were included in the meta-analysis: ten for interleukin (IL)-6, five for IL-8, three for IL-10, and ten for tumor necrosis factor alpha (TNF-α). Compared with controls, patients with narcolepsy had higher plasma levels of IL-6 (95% CI [0.22, 3.74]; P = 0.03) and TNF-α (95% CI [0.53, 4.18]; P = 0.01), while did not significantly differ in plasma IL-8 (95% CI [-1.64, 2.08]; P = 0.82) and IL-10 (95% CI [-1.29, 0.72]; P = 0.57) as well as serum IL-6 (95% CI [-1.48, 0.32], P = 0.21) and TNF-α (95% CI [-3.14, 0.19], P = 0.08) and CSF IL-8 (95% CI [-1.16, 0.41]; P = 0.35) levels. Patients with narcolepsy exhibited lower CSF IL-6 (95% CI [-0.66, 0.06]; P = 0.02) levels comparing with controls. CONCLUSIONS: Patients with narcolepsy had elevated plasma levels of IL-6 and TNF-α and lower levels of CSF IL-6 than non-narcoleptic controls. Our results support the role of inflammation in the pathophysiology of narcolepsy. However, plasma levels of IL-8 and IL-10, serum levels of IL-6 and TNF-α and CSF IL-8 did not significantly differ between patients and controls.


Cytokines/metabolism , Narcolepsy/metabolism , Humans , Interleukin-10/blood , Interleukin-6/blood , Interleukin-6/cerebrospinal fluid , Interleukin-8/blood , Interleukin-8/cerebrospinal fluid , Narcolepsy/blood , Narcolepsy/cerebrospinal fluid , Tumor Necrosis Factor-alpha/blood
17.
CNS Neurol Disord Drug Targets ; 19(2): 142-147, 2020.
Article En | MEDLINE | ID: mdl-32148204

BACKGROUND: Endocannabinoids (ECs) modulate both excitatory and inhibitory components in the CNS. There is a growing body of evidence that shows ECs influence both hypothalamic orexinergic and histaminergic neurons involved in narcolepsy physiopathology. Therefore, ECs may influence sleep and sleep-wake cycle. OBJECTIVE: To evaluate EC levels in the CSF of untreated narcoleptic patients to test whether ECs are dysregulated in Narcolepsy Type 1 (NT1) and Type 2 (NT2). METHODS: We compared CSF Anandamide (AEA), 2-Arachidonoylglycerol (2-AG) and orexin in narcoleptic drug-naïve patients and in a sample of healthy subjects. RESULTS: We compared NT1 (n=6), NT2 (n=6), and healthy controls (n=6). We found significantly reduced AEA levels in NT1 patients compared to both NT2 and controls. No differences were found between AEA levels in NT2 versus controls and between 2-AG levels in all groups, although a trend toward a decrease in NT1 was evident. Finally, the CSF AEA level was related to CSF orexin levels in all subjects. CONCLUSION: We demonstrated that the EC system is dysregulated in NT1.


Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Narcolepsy/metabolism , Polyunsaturated Alkamides/metabolism , Adult , Case-Control Studies , Female , Humans , Male , Middle Aged , Orexins/metabolism , Pilot Projects , Rome , Sleep/physiology , Young Adult
18.
J Neurosci ; 39(47): 9435-9452, 2019 11 20.
Article En | MEDLINE | ID: mdl-31628177

Narcolepsy, characterized by excessive daytime sleepiness, is associated with dysfunction of the hypothalamic hypocretin/orexin (Hcrt) system, either due to extensive loss of Hcrt cells (Type 1, NT1) or hypothesized Hcrt signaling impairment (Type 2, NT2). Accordingly, efforts to recapitulate narcolepsy-like symptoms in mice have involved ablating these cells or interrupting Hcrt signaling. Here, we describe orexin/Arch mice, in which a modified archaerhodopsin-3 gene was inserted downstream of the prepro-orexin promoter, resulting in expression of the yellow light-sensitive Arch-3 proton pump specifically within Hcrt neurons. Histological examination along with ex vivo and in vivo electrophysiological recordings of male and female orexin/Arch mice demonstrated silencing of Hcrt neurons when these cells were photoilluminated. However, high expression of the Arch transgene affected cellular and physiological parameters independent of photoillumination. The excitability of Hcrt neurons was reduced, and both circadian and metabolic parameters were perturbed in a subset of orexin/Arch mice that exhibited high levels of Arch expression. Orexin/Arch mice also had increased REM sleep under baseline conditions but did not exhibit cataplexy, a sudden loss of muscle tone during wakefulness characteristic of NT1. These aberrations resembled some aspects of mouse models with Hcrt neuron ablation, yet the number of Hcrt neurons in orexin/Arch mice was not reduced. Thus, orexin/Arch mice may be useful to investigate Hcrt system dysfunction when these neurons are intact, as is thought to occur in narcolepsy without cataplexy (NT2). These results also demonstrate the utility of extended phenotypic screening of transgenic models when specific neural circuits have been manipulated.SIGNIFICANCE STATEMENT Optogenetics has become an invaluable tool for functional dissection of neural circuitry. While opsin expression is often achieved by viral injection, stably integrated transgenes offer some practical advantages. Here, we demonstrate successful transgenic expression of an inhibitory opsin in hypocretin/orexin neurons, which are thought to promote or maintain wakefulness. Both brief and prolonged illumination resulted in inhibition of these neurons and induced sleep. However, even in the absence of illumination, these cells exhibited altered electrical characteristics, particularly when transgene expression was high. These aberrant properties affected metabolism and sleep, resulting in a phenotype reminiscent of the narcolepsy Type 2, a sleep disorder for which no good animal model currently exists.


Archaeal Proteins/biosynthesis , Brain/metabolism , Narcolepsy/metabolism , Neurons/metabolism , Orexins/metabolism , Animals , Archaeal Proteins/genetics , Brain/cytology , Brain Chemistry/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Narcolepsy/genetics , Neurons/chemistry , Optogenetics/methods , Orexins/genetics , Organ Culture Techniques
19.
Neuroimage Clin ; 23: 101875, 2019.
Article En | MEDLINE | ID: mdl-31174102

The aim of this study was to investigate structural changes in the brain stem of adolescents with narcolepsy, a disorder characterized by excessive daytime sleepiness, fragmented night-time sleep, and cataplexy. For this purpose, we used quantitative magnetic resonance imaging to obtain R1 and R2 relaxation rates, proton density, and myelin maps in adolescents with narcolepsy (n = 14) and healthy controls (n = 14). We also acquired resting state functional magnetic resonance imaging (fMRI) for brainstem connectivity analysis. We found a significantly lower R2 in the rostral reticular formation near the superior cerebellar peduncle in narcolepsy patients, family wise error corrected p = .010. Narcolepsy patients had a mean R2 value of 1.17 s-1 whereas healthy controls had a mean R2 of 1.31 s-1, which was a large effect size with Cohen d = 4.14. We did not observe any significant differences in R1 relaxation, proton density, or myelin content. The sensitivity of R2 to metal ions in tissue and the transition metal ion chelating property of neuromelanin indicate that the R2 deviant area is one of the neuromelanin containing nuclei of the brain stem. The close proximity and its demonstrated involvement in sleep-maintenance, specifically through orexin projections from the hypothalamus regulating sleep stability, as well as the results from the connectivity analysis, suggest that the observed deviant area could be the locus coeruleus or other neuromelanin containing nuclei in the proximity of the superior cerebellar peduncle. Hypothetically, the R2 differences described in this paper could be due to lower levels of neuromelanin in this area of narcolepsy patients.


Melanins , Narcolepsy/pathology , Reticular Formation/pathology , Adolescent , Female , Humans , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Male , Melanins/metabolism , Narcolepsy/metabolism , Neuroimaging/methods , Reticular Formation/metabolism , Young Adult
20.
Brain Res ; 1718: 212-222, 2019 09 01.
Article En | MEDLINE | ID: mdl-30998932

Narcolepsy (NA) is a primary sleep disorder characterized by loss of hypocretinergic/orexinergic neurons. NA is associated with an increased risk for metabolic disorders such as diabetes mellitus (DM). Proposed mechanisms for this association are alterations in food intake, disruption of energy balance, glucose tolerance, and insulin sensitivity, as well as inflammation and genetic factors. Orexin deficiency, is associated with increased food intake and reduced basal metabolic rate (BMR) both leading to obesity. The anti-apoptotic effect of orexin on pancreatic beta-cells, increase in peripheral insulin sensitivity, and reduced lipolysis in the adipose tissue, together confer an increased risk for obesity and type 2 DM (T2DM) in NA patients. The main pathomechanisms relating type 1 DM (T1DM) to NA involve autoimmunity and inflammation. HLA genes that confer a risk for NA, such as DQB1*0602 are protective against T1DM, while catepsin gene (CTSH) mutations are a risk factor for both NA and T1DM. Gestational DM (GDM) is associated with obesity which is a potential outcome of narcolepsy. GDM patients have lower serum orexin expression which is associated with increased fasting glucose and decreased fasting insulin. Ongoing research on the use of orexin receptor (OXR) antagonists in sleep disorders has opened a window to the pathomechanisms of NA and the potentials for OXR modulation in eating disorders and obesity. Understanding the common pathophysiological mechanisms of NA, DM and obesity could guide us in designing life-style modification programs, genetic consultations, and targeted therapies, such as immunotherapy, for obesity in NA.


Diabetes Mellitus/physiopathology , Narcolepsy/physiopathology , Orexins/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus/metabolism , Energy Metabolism , Humans , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Narcolepsy/metabolism , Neurons/metabolism , Obesity/physiopathology
...