Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Research Article

CSF Levels of the Endocannabinoid Anandamide are Reduced in Patients with Untreated Narcolepsy Type 1: A Pilot Study

Author(s): Andrea Romigi*, Monica Bari, Claudio Liguori, Francesca Izzi, Cinzia Rapino, Marzia Nuccetelli, Natalia Battista, Sergio Bernardini, Diego Centonze, Nicola Biagio Mercuri, Fabio Placidi and Mauro Maccarrone

Volume 19, Issue 2, 2020

Page: [142 - 147] Pages: 6

DOI: 10.2174/1871527319666200309115602

Price: $65

Abstract

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.

Keywords: Endocannabinoid, narcolepsy type 1, narcolepsy type 2, anandamide, 2-arachidonoylglycerol, cerebrospinal fluid.

Graphical Abstract
[1]
Scammell TE. Narcolepsy. N Engl J Med 2015; 373(27): 2654-62.
[http://dx.doi.org/10.1056/NEJMra1500587] [PMID: 26716917]
[2]
Arias-Carrion O, Ortega-Robles E, de Celis-Alonso B, et al. Depletion of hypocretin/orexin neurons increases cell proliferation in the adult subventricular zone. CNS Neurol Disord Drug Targets 2018; 17(2): 106-12.
[http://dx.doi.org/10.2174/1871527317666180314115623] [PMID: 29542425]
[3]
Chiurchiù V, van der Stelt M, Centonze D, Maccarrone M. The endocannabinoid system and its therapeutic exploitation in multiple sclerosis: Clues for other neuroinflammatory diseases. Prog Neurobiol 2018; 160: 82-100.
[http://dx.doi.org/10.1016/j.pneurobio.2017.10.007] [PMID: 29097192]
[4]
Di Marzo V. Targeting the endocannabinoid system: to enhance or reduce? Nat Rev Drug Discov 2008; 7(5): 438-55.
[http://dx.doi.org/10.1038/nrd2553] [PMID: 18446159]
[5]
Friedman D, French JA, Maccarrone M. Safety, efficacy, and mechanisms of action of cannabinoids in neurological disorders. Lancet Neurol 2019; 18: 504-12.
[6]
Prospéro-García O, Amancio-Belmont O, Becerril Meléndez AL, Ruiz-Contreras AE, Méndez-Díaz M. Endocannabinoids and sleep. Neurosci Biobehav Rev 2016; 71: 671-9.
[http://dx.doi.org/10.1016/j.neubiorev.2016.10.005] [PMID: 27756691]
[7]
Lunardelli ML, Crupi R, Siracusa R, et al. Co-ultraPEALut: role in preclinical and clinical delirium manifestations. CNS Neurol Disord Drug Targets 2019; 18(7): 530-54.
[8]
Onesti E, Frasca V, Ceccanti M, et al. Short-term ultramicronized palmitoylethanolamide therapy in patients with myasthenia gravis: a pilot study to possible future implications of treatment. CNS Neurol Disord Drug Targets 2019; 18(3): 232-8.
[http://dx.doi.org/10.2174/1871527318666190131121827] [PMID: 30706796]
[9]
Naganuma F, Bandaru SS, Absi G, Mahoney CE, Scammell TE, Vetrivelan R. Melanin-concentrating hormone neurons contribute to dysregulation of rapid eye movement sleep in narcolepsy. Neurobiol Dis 2018; 120: 12-20.
[http://dx.doi.org/10.1016/j.nbd.2018.08.012] [PMID: 30149182]
[10]
Murillo-Rodríguez E, Millán-Aldaco D, Palomero-Rivero M, Morales-Lara D, Mechoulam R, Drucker-Colín R. Cannabidiol partially blocks the sleepiness in hypocretin-deficient rats. Preliminary data. CNS Neurol Disord Drug Targets 2019.
[11]
Liguori C, Placidi F, Izzi F, et al. Beta-amyloid and phosphorylated tau metabolism changes in narcolepsy over time. Sleep Breath 2016; 20(1): 277-83.
[http://dx.doi.org/10.1007/s11325-015-1305-9] [PMID: 26803606]
[12]
Liguori C, Placidi F, Izzi F, et al. May CSF beta-amyloid and tau proteins levels be influenced by long treatment duration and stable medication in narcolepsy? Sleep Med 2014; 15(11): 1424.
[http://dx.doi.org/10.1016/j.sleep.2014.07.002] [PMID: 25192675]
[13]
Liguori C, Placidi F, Albanese M, et al. CSF beta-amyloid levels are altered in narcolepsy: a link with the inflammatory hypothesis? J Sleep Res 2014; 23(4): 420-4.
[http://dx.doi.org/10.1111/jsr.12130] [PMID: 24635662]
[14]
Romigi A, Bari M, Placidi F, et al. Cerebrospinal fluid levels of the endocannabinoid anandamide are reduced in patients with untreated newly diagnosed temporal lobe epilepsy. Epilepsia 2010; 51(5): 768-72.
[http://dx.doi.org/10.1111/j.1528-1167.2009.02334.x] [PMID: 19817812]
[15]
Ono T, Kanbayashi T, Yoshizawa K, Nishino S, Shimizu T. Measurement of cerebrospinal fluid orexin-A (hypocretin-1) by enzyme-linked immunosorbent assay: A comparison with radioimmunoassay. Psychiatry Clin Neurosci 2018; 72(11): 849-50.
[http://dx.doi.org/10.1111/pcn.12780] [PMID: 30159979]
[16]
Liguori C, Moresco M, Izzi F, Mercuri NB, Plazzi G, Placidi F. New revolution in the assessment of cerebrospinal fluid orexin-A: Enzyme-linked immunosorbent assay! Psychiatry Clin Neurosci 2019; 73(4): 194-5.
[http://dx.doi.org/10.1111/pcn.12816] [PMID: 30588717]
[17]
Liguori C, Romigi A, Nuccetelli M, et al. Orexinergic system dysregulation, sleep impairment, and cognitive decline in Alzheimer disease. JAMA Neurol 2014; 71(12): 1498-505.
[http://dx.doi.org/10.1001/jamaneurol.2014.2510] [PMID: 25322206]
[18]
Murillo-Rodríguez E, Morales-Lara D, Pastrana-Trejo JC, Macías-Triana L, Romero-Cordero K, de-la-Cruz M, et al. Cannabinoids, sleep, and the MCH system Melanin-Concentrating Hormone and Sleep. Cham: Springer International Publishing 2018; pp. 121-30.
[http://dx.doi.org/10.1007/978-3-319-75765-0_6]
[19]
Pérez-Morales M, De La Herrán-Arita AK, Méndez-Díaz M, Ruiz-Contreras AE, Drucker-Colín R, Prospéro-García O. 2-AG into the lateral hypothalamus increases REM sleep and cFos expression in melanin concentrating hormone neurons in rats. Pharmacol Biochem Behav 2013; 108: 1-7.
[http://dx.doi.org/10.1016/j.pbb.2013.04.006] [PMID: 23603032]
[20]
Ferreira JGP, Bittencourt JC, Adamantidis A. Melanin-concentrating hormone and sleep. Curr Opin Neurobiol 2017; 44: 152-8.
[http://dx.doi.org/10.1016/j.conb.2017.04.008] [PMID: 28527391]
[21]
Thompson MD, Sakurai T, Rainero I, Maj MC, Kukkonen JP. Orexin receptor multimerization versus functional interactions: neuropharmacological implications for opioid and cannabinoid signalling and pharmacogenetics. Pharmaceuticals (Basel) 2017; 10(4): E79
[http://dx.doi.org/10.3390/ph10040079] [PMID: 28991183]
[22]
Murillo-Rodriguez E, Pastrana-Trejo JC, Salas-Crisóstomo M, de-la-Cruz M. The endocannabinoid system modulating levels of consciousness, emotions and likely dream contents. CNS Neurol Disord Drug Targets 2017; 16(4): 370-9.
[http://dx.doi.org/10.2174/1871527316666170223161908] [PMID: 28240187]
[23]
Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non-psychotropic plant cannabinoids: new therapeutic opportunities from an ancient herb. Trends Pharmacol Sci 2009; 30(10): 515-27.
[http://dx.doi.org/10.1016/j.tips.2009.07.006] [PMID: 19729208]
[24]
Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol 2015; 172(20): 4790-805.
[http://dx.doi.org/10.1111/bph.13250] [PMID: 26218440]
[25]
Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol 2007; 150(5): 613-23.
[http://dx.doi.org/10.1038/sj.bjp.0707133] [PMID: 17245363]
[26]
Tham M, Yilmaz O, Alaverdashvili M, Kelly MEM, Denovan-Wright EM, Laprairie RB. Allosteric and orthosteric pharmacology of cannabidiol and cannabidiol-dimethylheptyl at the type 1 and type 2 cannabinoid receptors. Br J Pharmacol 2019; 176(10): 1455-69.
[http://dx.doi.org/10.1111/bph.14440] [PMID: 29981240]
[27]
Calpe-López C, García-Pardo MP, Aguilar MA. Cannabidiol treatment might promote resilience to cocaine and methamphetamine use disorders: A review of possible mechanisms. Molecules 2019; 24(14): E2583
[http://dx.doi.org/10.3390/molecules24142583] [PMID: 31315244]
[28]
Zlebnik NE, Cheer JF. Beyond the CB1 receptor: is cannabidiol the answer for disorders of motivation? Annu Rev Neurosci 2016; 39: 1-17.
[http://dx.doi.org/10.1146/annurev-neuro-070815-014038] [PMID: 27023732]
[29]
Leweke FM, Piomelli D, Pahlisch F, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry 2012; 2: e94
[http://dx.doi.org/10.1038/tp.2012.15] [PMID: 22832859]
[30]
Campos AC, Ortega Z, Palazuelos J, et al. The anxiolytic effect of cannabidiol on chronically stressed mice depends on hippocampal neurogenesis: involvement of the endocannabinoid system. Int J Neuropsychopharmacol 2013; 16(6): 1407-19.
[http://dx.doi.org/10.1017/S1461145712001502] [PMID: 23298518]
[31]
Pisanti S, Malfitano AM, Ciaglia E, et al. Cannabidiol: State of the art and new challenges for therapeutic applications. Pharmacol Ther 2017; 175: 133-50.
[http://dx.doi.org/10.1016/j.pharmthera.2017.02.041] [PMID: 28232276]
[32]
Martinotti G, Santacroce R, Papanti D, Elgharably Y, Prilutskaya M, Corazza O. Synthetic cannabinoids: psychopharmacology, clinical aspects, psychotic onset. CNS Neurol Disord Drug Targets 2017; 16(5): 567-75.
[http://dx.doi.org/10.2174/1871527316666170413101839] [PMID: 28412921]
[33]
Pichini S, Busardo FP. Editorial: cannabis: neurological correlates in abuse and medical use. CNS Neurol Disord Drug Targets 2017; 16(5): 524-6.
[http://dx.doi.org/10.2174/187152731605170810115956] [PMID: 28847308]
[34]
Solimini R, Rotolo MC, Pichini S, Pacifici R. Neurological disorders in medical use of cannabis: an update. CNS Neurol Disord Drug Targets 2017; 16(5): 527-33.
[http://dx.doi.org/10.2174/1871527316666170413105421] [PMID: 28412919]
[35]
Medeiros MRB, de Mello Alves Rodrigues AC, Alves MR, et al. Bibliometrics of CNS & neurological disorders - drug targets: an international evolution along time. CNS Neurol Disord Drug Targets 2019; 18(3): 239-44.
[http://dx.doi.org/10.2174/1871527318666181227123924] [PMID: 30588889]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy