Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Brain Behav ; 14(7): e3623, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38988104

ABSTRACT

INTRODUCTION: Multiple sclerosis (MS) is a debilitating neurological condition affecting nearly one million people across the United States. Among the most prominent symptoms of the condition are excessive fatigue and daytime sleepiness. Numerous clinical trials have investigated the efficacy of modafinil in addressing fatigue among these patients. OBJECTIVE: The objective of the present study is to assess the safety and efficacy of modafinil for the treatment of fatigue in MS. METHODOLOGY: An electronic search of PUBMED, ScienceDirect, and Cochrane Central was conducted for articles published from inception to December 2023 using search terms such as "modafinil," "fatigue," and "MS." RESULTS: Seven studies were included in our analysis. Modafinil leads to a meaningful reduction in fatigue when compared with placebo, as measured by Modified Fatigue Impact Scale [mean difference (MD) = -4.42 [-8.01, -.84]; I2 = 45%; p = .02] and Epworth Sleepiness Scale [MD = -.87 [-1.64, -.10]; I2 = 0%; p = .03]. Modafinil also demonstrated a greater risk of precipitating adverse events (e.g., insomnia, gastrointestinal symptoms) when compared with placebo [RR = 1.30 [1.03, 1.66]; I2 = 0%; p = .03]. In quality-of-life assessments, modafinil was associated with overall improvement in well-being [standardized mean difference = .18 [.01, .35]; I2 = 56%; p = .04]. CONCLUSION: The data indicates that modafinil confers a therapeutic benefit when treating fatigue in patients with MS and improves overall quality of life; however, there is a risk of precipitating adverse events. Ultimately, higher quality of evidence may be required to better inform clinical management.


Subject(s)
Fatigue , Modafinil , Multiple Sclerosis , Modafinil/therapeutic use , Modafinil/adverse effects , Modafinil/pharmacology , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/complications , Fatigue/drug therapy , Fatigue/etiology , Wakefulness-Promoting Agents/therapeutic use , Wakefulness-Promoting Agents/adverse effects , Wakefulness-Promoting Agents/pharmacology , Controlled Clinical Trials as Topic
2.
Sleep Med ; 119: 95-102, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38669836

ABSTRACT

BACKGROUND: Patients with narcolepsy often experience disturbed nighttime sleep. Modafinil is commonly prescribed for hypersomnolence, but its impacts on nocturnal sleep remain unclear. This study uses actigraphy to examine the effect of modafinil on both hypersomnolence and nocturnal sleep patterns in patients with narcolepsy. METHODS: Prior to treatment, 87 patients with narcolepsy wore an actigraphy for 7-14 days to assess their nighttime sleep. After evaluation, they received a daily dose of 200-400 mg of modafinil in the morning and wore an actigraphy again six months after initiating treatment. Questionnaires, including the Epworth-Sleepiness-Scale (ESS), the Visual-Analogue-for-Hypersomnolence (VAS), and the Short-Form-36-Health-Survey (SF-36), were used to evaluate hypersomnolence and quality of life both before and after treatment. Paired t-tests and independent samples t-tests were used for pre- and post-treatment comparisons and subgroup analysis. We used the Pearson's correlation test to measure the correlations between the sleep parameters of the actigraphy and data of the questionnaires. RESULTS: Improvements in hypersomnolence were noted following modafinil treatment, and we observed no significant deterioration in nocturnal sleep parameters by the actigraphy. The total number of awakenings by actigraphy significantly decreased (p = 0.005), especially in females (p = 0.008), while sleep onset latency significantly increased in children/adolescents (p = 0.014). Correlations were found between the sleep parameters of the actigraphy and ESS, VAS, and SF-36 scores. CONCLUSION: Modafinil treatment may not worsen nighttime sleep in patients with narcolepsy. However, it should be administered with care in children and adolescents.


Subject(s)
Actigraphy , Benzhydryl Compounds , Modafinil , Narcolepsy , Quality of Life , Wakefulness-Promoting Agents , Humans , Modafinil/therapeutic use , Modafinil/pharmacology , Narcolepsy/drug therapy , Female , Male , Benzhydryl Compounds/therapeutic use , Benzhydryl Compounds/pharmacology , Adult , Wakefulness-Promoting Agents/therapeutic use , Wakefulness-Promoting Agents/pharmacology , Adolescent , Cohort Studies , Surveys and Questionnaires , Middle Aged , Young Adult , Sleep/drug effects , Central Nervous System Stimulants/therapeutic use , Central Nervous System Stimulants/pharmacology , Child , Treatment Outcome
3.
Curr Neuropharmacol ; 22(11): 1899-1908, 2024.
Article in English | MEDLINE | ID: mdl-38486390

ABSTRACT

INTRODUCTION: Armodafinil is a psychostimulant that promotes alertness, and it has been shown to improve attention, memory, and fatigue in healthy adults and adults with neurodevelopmental conditions that share symptoms with Attention Deficit Hyperactivity Disorder (ADHD). It is generally well tolerated and safe, and most of the adverse events reported are considered not serious. However, the available evidence on the efficacy of armodafinil for the treatment of ADHD in adults is scarce. OBJECTIVE: The present review aims to perform a systematized search of the available evidence on the possible therapeutic benefit of armodafinil treatment in adult patients with ADHD. METHODS: A literature review using PubMed was conducted to compile and summarize the available clinical and scientific evidence on the possible use of armodafinil as a pharmacological treatment in adult patients with ADHD. RESULTS: From the 86 articles reviewed, the available evidence showed that both acute and chronic treatment with armodafinil can improve wakefulness, memory, impulse control, and executive functions in adults with sleep disorders and other conditions. In addition, evidence of improvement in cognitive functions and mood alterations in other neuropsychiatric conditions was shown. CONCLUSION: Armodafinil could be useful for the treatment of ADHD in adults, according to the review of the literature from both pre-clinical and clinical studies.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Modafinil , Humans , Modafinil/therapeutic use , Modafinil/pharmacology , Attention Deficit Disorder with Hyperactivity/drug therapy , Wakefulness-Promoting Agents/therapeutic use , Wakefulness-Promoting Agents/pharmacology , Adult , Animals , Central Nervous System Stimulants/therapeutic use , Central Nervous System Stimulants/pharmacology , Benzhydryl Compounds/therapeutic use , Benzhydryl Compounds/pharmacology
4.
Arch. bronconeumol. (Ed. impr.) ; 59(12): 805-812, dic. 2023. ilus, graf
Article in English | IBECS | ID: ibc-228400

ABSTRACT

Introduction: Obstructive sleep apnea (OSA) is a chronic condition characterized by intermittent hypoxia (IH) and sleep fragmentation (SF). OSA can induce excessive daytime sleepiness (EDS) and is associated with impaired cognition and anxiety. Solriamfetol (SOL) and modafinil (MOD) are widely used wake-promoting agents in OSA patients with EDS. Methods: Male C57Bl/6J mice were exposed to SF along with sleep controls (SC) or to IH and room air (RA) controls during the light (inactive) phase for 4 and 16 weeks, respectively. Both IH and SF exposures were then discontinued to mimic “ideal” continuous positive airway pressure (CPAP) adherence. All groups were then randomly assigned to receive once daily intraperitoneal injections of SOL, MOD, or vehicle (VEH) for 6 days. Sleep/wake activity was assessed along with tests of explicit memory, anxiety and depression were performed before and after treatments. Results: IH and SF exposures increased sleep percentage in the dark phase and reduced wake bouts lengths (i.e., EDS), and induced cognitive deficits and impulsivity in mice. Both SOL and MOD treatments effectively mitigated EDS when combined with recovery, while recovery alone did not improve EDS over the 6-day period. Furthermore, improvements explicit memory emerged only after SOL. Conclusion: Chronic IH and SF induce EDS in young adult mice that is not ameliorated by recovery except when combined with either SOL or MOD. SOL, but not MOD, significantly improves IH-induced cognitive deficits. Thus, SOL emerges as a viable adjuvant medication for residual EDS in OSA along with its positive impact on cognition. (AU)


Subject(s)
Animals , Mice , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/therapy , Disorders of Excessive Somnolence/etiology , Wakefulness-Promoting Agents/pharmacology , Wakefulness-Promoting Agents/therapeutic use , Modafinil/pharmacology , Modafinil/therapeutic use , Cognition , Hypoxia
5.
Arch Bronconeumol ; 59(12): 805-812, 2023 Dec.
Article in English, Spanish | MEDLINE | ID: mdl-37783638

ABSTRACT

INTRODUCTION: Obstructive sleep apnea (OSA) is a chronic condition characterized by intermittent hypoxia (IH) and sleep fragmentation (SF). OSA can induce excessive daytime sleepiness (EDS) and is associated with impaired cognition and anxiety. Solriamfetol (SOL) and modafinil (MOD) are widely used wake-promoting agents in OSA patients with EDS. METHODS: Male C57Bl/6J mice were exposed to SF along with sleep controls (SC) or to IH and room air (RA) controls during the light (inactive) phase for 4 and 16 weeks, respectively. Both IH and SF exposures were then discontinued to mimic "ideal" continuous positive airway pressure (CPAP) adherence. All groups were then randomly assigned to receive once daily intraperitoneal injections of SOL, MOD, or vehicle (VEH) for 6 days. Sleep/wake activity was assessed along with tests of explicit memory, anxiety and depression were performed before and after treatments. RESULTS: IH and SF exposures increased sleep percentage in the dark phase and reduced wake bouts lengths (i.e., EDS), and induced cognitive deficits and impulsivity in mice. Both SOL and MOD treatments effectively mitigated EDS when combined with recovery, while recovery alone did not improve EDS over the 6-day period. Furthermore, improvements explicit memory emerged only after SOL. CONCLUSION: Chronic IH and SF induce EDS in young adult mice that is not ameliorated by recovery except when combined with either SOL or MOD. SOL, but not MOD, significantly improves IH-induced cognitive deficits. Thus, SOL emerges as a viable adjuvant medication for residual EDS in OSA along with its positive impact on cognition.


Subject(s)
Disorders of Excessive Somnolence , Sleep Apnea, Obstructive , Wakefulness-Promoting Agents , Humans , Male , Animals , Mice , Wakefulness , Wakefulness-Promoting Agents/pharmacology , Wakefulness-Promoting Agents/therapeutic use , Continuous Positive Airway Pressure , Disease Models, Animal , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/therapy , Modafinil/pharmacology , Modafinil/therapeutic use , Disorders of Excessive Somnolence/etiology , Hypoxia , Cognition
6.
Pharmacol Res Perspect ; 9(5): e00855, 2021 10.
Article in English | MEDLINE | ID: mdl-34423920

ABSTRACT

Several therapeutic options are currently available to treat excessive daytime sleepiness (EDS) in patients suffering from narcolepsy or obstructive sleep apnea. However, there are no comparisons between the various wake-promoting agents in terms of mechanism of action, efficacy, or safety. The goal of this study was to compare amphetamine, modafinil, solriamfetol, and pitolisant at their known primary pharmacological targets, histamine H3 receptors (H3R), dopamine, norepinephrine, and serotonin transporters, and in various in vivo preclinical models in relation to neurochemistry, locomotion, behavioral sensitization, and food intake. Results confirmed that the primary pharmacological effect of amphetamine, modafinil, and solriamfetol was to increase central dopamine neurotransmission, in part by inhibiting its transporter. Furthermore, solriamfetol increased levels of extracellular dopamine in the nucleus accumbens, and decreased the 3,4-dihydroxyphenyl acetic acid (DOPAC)/DA ratio in the striatum, as reported for modafinil and amphetamine. All these compounds produced hyperlocomotion, behavioral sensitization, and hypophagia, which are common features of psychostimulants and of compounds with abuse potential. In contrast, pitolisant, a selective and potent H3R antagonist/inverse agonist that promotes wakefulness, had no effect on striatal dopamine, locomotion, or food intake. In addition, pitolisant, devoid of behavioral sensitization by itself, attenuated the hyperlocomotion induced by either modafinil or solriamfetol. Therefore, pitolisant presents biochemical, neurochemical, and behavioral profiles different from those of amphetamine and other psychostimulants such as modafinil or solriamfetol. In conclusion, pitolisant is a differentiated therapeutic option, when compared with psychostimulants, for the treatment of EDS, as this agent does not show any amphetamine-like properties within in vivo preclinical models.


Subject(s)
Amphetamine/pharmacology , Carbamates/pharmacology , Corpus Striatum/drug effects , Disorders of Excessive Somnolence/drug therapy , Feeding Behavior/drug effects , Locomotion/drug effects , Modafinil/pharmacology , Phenylalanine/analogs & derivatives , Piperidines/pharmacology , Wakefulness-Promoting Agents/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Corpus Striatum/metabolism , Disorders of Excessive Somnolence/etiology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Drug Evaluation, Preclinical , Drug Inverse Agonism , Histamine Antagonists/pharmacology , Mice , Narcolepsy/drug therapy , Neostriatum/drug effects , Neostriatum/metabolism , Norepinephrine Plasma Membrane Transport Proteins/drug effects , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phenylalanine/pharmacology , Receptors, Histamine H3 , Sleep Apnea, Obstructive/complications
7.
Pharmacol Biochem Behav ; 196: 172968, 2020 09.
Article in English | MEDLINE | ID: mdl-32593791

ABSTRACT

BACKGROUND: Individual responses to the effects of inadequate sleep have been well documented; some people are more vulnerable to the effects of sleep loss than others. Fatigue-vulnerable individuals generally require access to effective fatigue countermeasures; however, the question arises as to whether these fatigue-vulnerable individuals receive the same benefits shown in group efficacy data. The present study administered modafinil to individuals to determine its differential effects on performance of best and worst performers during sleep deprivation. METHODS: A sample of 22 men, age 21-40 yrs., was tested on 2 separate occasions during which they were kept awake for 36 h. During one period they received 200 mg modafinil; during the other they received placebo. Participants were tested on a variety of tasks while rested and at 5-hr intervals across the continuous wakefulness period. Performance for each cognitive task and subjective measure of fatigue from the placebo period was used to group individuals into high (HP) or low performance (LP) groups to indicate fatigue vulnerability for each task. RESULTS: Results indicated that on the MTS task, the HP group performed the same throughout the testing period, regardless of whether they received modafinil or not. However, the LP group significantly improved after receiving modafinil compared to placebo. Performance on the PVT showed the HP group had a small decrease in the number of lapses after receiving modafinil compared to placebo, whereas the LP group had a large decrease in lapses after receiving modafinil compared to placebo. Performance on the RDM showed no difference between groups, regardless of drug condition. Groups did not differ after receiving modafinil on subjective fatigue measured by the POMS. CONCLUSIONS: Depending on the task, HP individuals did not benefit substantially when administered modafinil compared to placebo. However, the LP individuals improved after receiving modafinil compared to placebo.


Subject(s)
Modafinil/pharmacology , Sleep Deprivation/physiopathology , Wakefulness-Promoting Agents/pharmacology , Adult , Double-Blind Method , Humans , Male , Placebos , Psychomotor Performance/drug effects , Reaction Time/drug effects , Young Adult
8.
Neurotox Res ; 38(2): 498-507, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32367472

ABSTRACT

Psychostimulant drugs, such as modafinil and caffeine, induce transcriptional alterations through the dysregulation of epigenetic mechanisms. We have previously demonstrated that acute modafinil administration is accompanied by multiple changes in the expression of histone deacetylases (HDACs) within the mouse medial prefrontal cortex (mPFC). Herein, we compared alterations in class IIa HDACs in the mouse mPFC and dorsal striatum (DS) after a single exposure to each psychostimulant. We treated male C57BL/6 mice with modafinil (90 mg/kg, i.p.), caffeine (10 mg/kg, i.p.), or vehicle and evaluated locomotor activity. Following, we examined hdac4, hdac5, and hdac7 mRNA expression using qRT-PCR and HDAC7, pHDAC7, and pHDACs4/5/7 using Western blot. Last, we explored generalized effects in N2a cell line using modafinil (100 µM and 1 mM) or caffeine (80 µM and 800 µM). Our results indicate that modafinil had greater effects on locomotor activity compared with caffeine. qRT-PCR experiments revealed that modafinil decreased hdac5 and hdac7 mRNA expression in the DS, while caffeine had no effects. In the mPFC, modafinil increased hdac7 mRNA expression, with no effects observed for caffeine. Western blot revealed that within the DS, modafinil induced increases in HDAC7, pHDAC7, and pHDACs4/5/7 protein expression, while, in the mPFC, caffeine induced decreases in HDAC7, pHDAC7, and pHDACs4/5/7 protein levels. In vitro studies revealed that modafinil increased hdac4, hdac5, and hdac7 mRNA levels in N2a, while caffeine only increased hdac5 at a higher dose. These findings support the notion that modafinil and caffeine exert distinct regulation of class IIa HDAC family members and that these transcriptional and translational consequences are region-specific.


Subject(s)
Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Histone Deacetylases/drug effects , Locomotion/drug effects , Modafinil/pharmacology , Animals , Cell Line , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Wakefulness-Promoting Agents/pharmacology
9.
Med Hypotheses ; 134: 109434, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31634772

ABSTRACT

Dexamethasone has been observed to cause night-time wakefulness without accompanying fatigue. It is proposed here that the drug interrupts a sleep signal, and that although sleep has an ultimate ecological function, this signal is a proximate mechanism but not an ultimate physiological necessity, such that sleep is not a necessity.


Subject(s)
Models, Neurological , Sleep/physiology , Adaptation, Physiological , Cytokines/physiology , Dexamethasone/pharmacology , Fatigue/drug therapy , Fatigue/physiopathology , Humans , Sleep/drug effects , Sleep Deprivation/physiopathology , Wakefulness/physiology , Wakefulness-Promoting Agents/pharmacology , Wakefulness-Promoting Agents/therapeutic use , gamma-Aminobutyric Acid/physiology
10.
Pain ; 161(2): 288-299, 2020 02.
Article in English | MEDLINE | ID: mdl-31651580

ABSTRACT

Patients with chronic pain often report being sensitive to pain at night before falling asleep, a time when the synchronization of cortical activity is initiated. However, how cortical activity relates to pain sensitivity is still unclear. Because sleep is characterized by enhanced cortical delta power, we hypothesized that enhanced cortical delta power may be an indicator of intensified pain. To test this hypothesis, we used pain thresholds tests, EEG/electromyogram recordings, c-Fos staining, and chemogenetic and pharmacological techniques in mice. We found that sleep deprivation or pharmacologic enhancement of EEG delta power by reserpine and scopolamine dramatically decreased mechanical pain thresholds, but not thermal withdrawal latency, in a partial sciatic nerve ligation model of neuropathic pain mice. On the contrary, suppression of EEG delta power using a wake-promoting agent modafinil significantly attenuated mechanical allodynia. Moreover, when EEG delta power was enhanced, c-Fos expression decreased in most regions of the cortex, except the anterior cingulate cortex (ACC), where c-Fos was increased in the somatostatin- and parvalbumin-positive GABAergic neurons. Chemogenetic activation of GABAergic neurons in ACC enhanced EEG delta power and lowered mechanical pain thresholds simultaneously in naive mice. However, chemogenetic inhibition of ACC GABAergic neurons could not block mechanical allodynia. These results provided compelling evidence that elevated EEG delta power is accompanied with aggravated neuropathic pain, whereas decreased delta power attenuated it, suggesting that enhanced delta power can be a specific marker of rising chronic neuropathic pain and that wake-promoting compounds could be used as analgesics in the clinic.


Subject(s)
Cerebral Cortex/physiopathology , Delta Rhythm/physiology , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Pain Threshold/physiology , Sleep/physiology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinergic Antagonists/pharmacology , Cortical Synchronization/drug effects , Cortical Synchronization/physiology , Delta Rhythm/drug effects , Electroencephalography , Electromyography , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Gyrus Cinguli/drug effects , Gyrus Cinguli/metabolism , Hyperalgesia/metabolism , Mice , Modafinil/pharmacology , Neuralgia/metabolism , Pain Threshold/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Reserpine/pharmacology , Sciatic Nerve/surgery , Scopolamine/pharmacology , Sleep/drug effects , Sleep Deprivation/chemically induced , Sleep Deprivation/physiopathology , Wakefulness-Promoting Agents/pharmacology
11.
Br J Clin Pharmacol ; 85(11): 2623-2633, 2019 11.
Article in English | MEDLINE | ID: mdl-31419329

ABSTRACT

AIMS: THN102 is a novel combination of modafinil and low-dose flecainide, targeting glial connexin activity to modulate modafinil effects. We investigated THN102 efficacy compared to modafinil and to placebo on vigilance and cognitive function during 40-hour total sleep deprivation (TSD). METHODS: Twenty healthy men participated in a double-blind, randomized, incomplete-block 3-period cross-over trial with 5 treatments (n = 12 per group): placebo (PBO), modafinil 100 mg (MOD100), THN102 100/1, 100/3, 100/9 (modafinil 100 mg and flecainide 1, 3 or 9 mg). Each period included a baseline day and a TSD day with treatments administered 3 times (01:00, 09:00 and 19:00). Reaction time in psychomotor vigilance test, subjective somnolence and vital signs were assessed before and during treatment. Working memory (2-Back) and executive processes (Go/noGo for vigilance and inhibition, Wisconsin card sorting task for mental flexibility, and Tower of London test for planning) were evaluated at 16:30. RESULTS: At 5 hours postdose−1 (after 23 hours TSD, primary endpoint), THN102 100/1 resulted in statistically higher psychomotor vigilance test speed vs MOD100 (3.97 ± 0.09 vs 3.74 ± 0.14, P < .05). No increase in effect was observed with higher flecainide doses in combinations. Most THN102 doses vs MOD100 also improved the number of correct responses in 2-Back and Go errors in Go/noGo (P < .05 for all doses), and perseverative responses in Wisconsin card sorting task (for 100/1 and 100/9). No impact on cardiac conduction was noted with THN102, and safety was similar to MOD100. CONCLUSIONS: THN102 seems more efficient than modafinil on vigilance, working memory and executive functions, opening new perspectives in management of hypersomnolence disorders.


Subject(s)
Flecainide/pharmacology , Modafinil/pharmacology , Neuroglia/drug effects , Sleep Deprivation/drug therapy , Wakefulness-Promoting Agents/pharmacology , Adult , Arousal/drug effects , Cognition/drug effects , Connexins/antagonists & inhibitors , Cross-Over Studies , Drug Combinations , Flecainide/therapeutic use , Healthy Volunteers , Humans , Male , Memory, Short-Term/drug effects , Modafinil/therapeutic use , Neuroglia/metabolism , Psychomotor Performance/drug effects , Reaction Time/drug effects , Treatment Outcome , Wakefulness/drug effects , Wakefulness-Promoting Agents/therapeutic use , Young Adult
12.
Clin Neurophysiol ; 130(9): 1488-1498, 2019 09.
Article in English | MEDLINE | ID: mdl-31295717

ABSTRACT

OBJECTIVE: It has been reported that sleep deprivation affects the neurophysiological mechanisms underpinning the vigilance. Here, we tested the following hypotheses in the PharmaCog project (www.pharmacog.org): (i) sleep deprivation may alter posterior cortical delta and alpha sources of resting state eyes-closed electroencephalographic (rsEEG) rhythms in healthy young adults; (ii) after the sleep deprivation, a vigilance enhancer may recover those rsEEG source markers. METHODS: rsEEG data were recorded in 36 healthy young adults before (Pre-sleep deprivation) and after (Post-sleep deprivation) one night of sleep deprivation. In the Post-sleep deprivation, these data were collected after a single dose of PLACEBO or MODAFINIL. rsEEG cortical sources were estimated by eLORETA freeware. RESULTS: In the PLACEBO condition, the sleep deprivation induced an increase and a decrease in posterior delta (2-4 Hz) and alpha (8-13 Hz) source activities, respectively. In the MODAFINIL condition, the vigilance enhancer partially recovered those source activities. CONCLUSIONS: The present results suggest that posterior delta and alpha source activities may be both related to the regulation of human brain arousal and vigilance in quiet wakefulness. SIGNIFICANCE: Future research in healthy young adults may use this methodology to preselect new symptomatic drug candidates designed to normalize brain arousal and vigilance in seniors with dementia.


Subject(s)
Brain Waves/drug effects , Cerebral Cortex/drug effects , Modafinil/pharmacology , Rest/physiology , Sleep Deprivation/physiopathology , Wakefulness-Promoting Agents/pharmacology , Adult , Alpha Rhythm/drug effects , Alpha Rhythm/physiology , Beta Rhythm/drug effects , Beta Rhythm/physiology , Brain Waves/physiology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/physiology , Cross-Over Studies , Delta Rhythm/drug effects , Delta Rhythm/physiology , Electroencephalography/methods , Functional Laterality , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Healthy Volunteers , Humans , Male , Sample Size , Theta Rhythm/drug effects , Theta Rhythm/physiology , Wakefulness/drug effects , Wakefulness/physiology
13.
Neuroscience ; 413: 86-98, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31202706

ABSTRACT

Glutamate is the major excitatory neurotransmitter in the brain and plays an essential role in regulating wakefulness. Histaminergic neurons, which are exclusively localized in the tuberomammillary nucleus (TMN) of the hypothalamus, have a pivotal role in the regulation of sleep-wake patterns by sending widespread projections into many brain areas implicated in sleep-wake control. The role of glutamate in histaminergic neurons within the TMN and the resulting sleep-wake profile remains unknown. We found that glutamate, NMDA, AMPA or dihydrokainate, a glutamate-uptake inhibitor, dose-dependently increased wakefulness when microinjected into the rat TMN. Glutamate, NMDA, and AMPA also increased the firing rate of action potentials in TMN histaminergic neurons. The arousal-promoting effect of glutamate was inhibited by NMDA and histamine H1 receptor antagonists. Furthermore, MK-801, an NMDA receptor antagonist, inhibited the firing rate of histaminergic neurons and increased non-rapid eye movement sleep after microinjection into rat TMN. Taken together, these findings demonstrated that glutamate activated histaminergic neurons in the TMN and increased wakefulness in rats, possibly via the action of NMDA and histamine H1 receptors.


Subject(s)
Glutamic Acid/pharmacology , Hypothalamic Area, Lateral/drug effects , Receptors, Histamine/metabolism , Wakefulness-Promoting Agents/pharmacology , Wakefulness/drug effects , Action Potentials/drug effects , Animals , Histamine H1 Antagonists/pharmacology , Hypothalamic Area, Lateral/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Sleep/drug effects , Sleep/physiology , Tissue Culture Techniques , Wakefulness/physiology
14.
BMC Neurosci ; 20(1): 14, 2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30894126

ABSTRACT

BACKGROUND: Previous data show that serotonin 2C (5-HT2C) and cannabinoid 1 (CB1) receptors have a role in the modulation of sleep-wake cycle. Namely, antagonists on these receptors promoted wakefulness and inhibited rapid eye movement sleep (REMS) in rodents. The interaction of these receptors are also present in other physiological functions, such as the regulation of appetite. Blockade of 5-HT2C receptors modulat the effect of CB1 receptor antagonist, presumably in consecutive or interdependent steps. Here we investigate, whether previous blockade of 5-HT2C receptors can affect CB1 receptor functions in the sleep-wake regulation. RESULTS: Wistar rats were equipped with electroencephalography (EEG) and electromyography (EMG) electrodes. Following the recovery and habituation after surgery, animals were injected intraperitoneally (ip.) with SB-242084, a 5-HT2C receptor antagonist (1.0 mg/kg) at light onset (beginning of passive phase) followed by an injection with AM-251, a CB1 receptor antagonist (5.0 or 10.0 mg/kg, ip.) 10 min later. EEG, EMG and motor activity were analyzed for the subsequent 2 h. Both SB-242084 and AM-251 increased the time spent in active wakefulness, while decreased the time spent in non-REMS and REMS stages in the first 2 h of passive phase. In combination, the effect of the agents were additive, furthermore, statistical analysis did not show any interaction between the effects of these drugs in the modulation of vigilance stages. CONCLUSIONS: Our results suggest that 5-HT2C receptor blockade followed by blockade of CB1 receptors evoked additive effect on the regulation of sleep-wake pattern.


Subject(s)
Cannabinoid Receptor Antagonists/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Sleep/drug effects , Wakefulness-Promoting Agents/pharmacology , Wakefulness/drug effects , Aminopyridines/pharmacology , Animals , Drug Synergism , Electroencephalography , Electromyography , Indoles/pharmacology , Male , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Sleep/physiology , Wakefulness/physiology
15.
Sleep ; 42(5)2019 05 01.
Article in English | MEDLINE | ID: mdl-30722053

ABSTRACT

Acute chemogenetic inhibition of histamine (HA) neurons in adult mice induced nonrapid eye movement (NREM) sleep with an increased delta power. By contrast, selective genetic lesioning of HA neurons with caspase in adult mice exhibited a normal sleep-wake cycle overall, except at the diurnal start of the lights-off period, when they remained sleepier. The amount of time spent in NREM sleep and in the wake state in mice with lesioned HA neurons was unchanged over 24 hr, but the sleep-wake cycle was more fragmented. Both the delayed increase in wakefulness at the start of the night and the sleep-wake fragmentation are similar phenotypes to histidine decarboxylase knockout mice, which cannot synthesize HA. Chronic loss of HA neurons did not affect sleep homeostasis after sleep deprivation. However, the chronic loss of HA neurons or chemogenetic inhibition of HA neurons did notably reduce the ability of the wake-promoting compound modafinil to sustain wakefulness. Thus, part of modafinil's wake-promoting actions arise through the HA system.


Subject(s)
Histamine/genetics , Modafinil/therapeutic use , Neurons/drug effects , Sleep Deprivation/genetics , Wakefulness-Promoting Agents/therapeutic use , Wakefulness/drug effects , Animals , Electroencephalography/drug effects , Electroencephalography/methods , Genetic Vectors/administration & dosage , Histamine/deficiency , Homeostasis/drug effects , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Modafinil/pharmacology , Neurons/physiology , Sleep/drug effects , Sleep/physiology , Sleep Deprivation/drug therapy , Sleep Deprivation/metabolism , Wakefulness/physiology , Wakefulness-Promoting Agents/pharmacology
16.
Sleep Med ; 55: 1-5, 2019 03.
Article in English | MEDLINE | ID: mdl-30735912

ABSTRACT

STUDY OBJECTIVES: To assess the relationship between real and simulated driving performance and the objective level of alertness as measured by the Maintenance of Wakefulness Test (MWT) in patients suffering from narcolepsy or idiopathic hypersomnia. METHODS: Twenty-seven patients (10 patients with narcolepsy, type 1 (n = 7) and type 2 (n = 3), and 17 patients with idiopathic hypersomnia, mean age = 33.8 ± 11.1 years, range = 18-65 y; four males) were recruited in a randomized, crossover, double-blind placebo-controlled trial, and compared to 27 matched healthy controls. Patients were randomly assigned to receive modafinil (400 mg) or placebo before the driving test (2 h of real and 2 h of simulated highway driving for each patient). Standard deviation of lateral position (SDLP) of the vehicle in real and simulated driving and mean sleep latency in a 4 × 40 min MWT were assessed. RESULTS: Untreated patients presented shorter sleep latencies on the MWT (20.8 (IQ range 16.1-32.9) vs. 34.9 min (IQ range 28.1-40.0)) and worse simulated driving performance (P < 0.001) than treated patients. Nevertheless, treated patients still exhibited shorter mean sleep latencies on the MWT than controls (34.9 (IQ range 28.1-40.0) vs. 40 min (IQ range 37.1-40.0), P < 0.05), but driving performance was identical in both groups. The SDLP of the vehicle in real driving conditions and the MWT score correlated with the SDLP in simulated driving (respectively, r = 0.34, P < 0.05 and r = -0.56, P < 0.001). CONCLUSIONS: In patients with narcolepsy/idiopathic hypersomnia, simulated driving and MWT explore different dimensions of fitness-to-drive and could be used complementarily to better evaluate sleep-related driving impairment.


Subject(s)
Automobile Driving , Computer Simulation , Idiopathic Hypersomnia/diagnosis , Narcolepsy/diagnosis , Wakefulness/physiology , Adult , Automobile Driving/psychology , Cross-Over Studies , Double-Blind Method , Humans , Idiopathic Hypersomnia/drug therapy , Idiopathic Hypersomnia/psychology , Middle Aged , Modafinil/pharmacology , Modafinil/therapeutic use , Narcolepsy/drug therapy , Narcolepsy/psychology , Wakefulness/drug effects , Wakefulness-Promoting Agents/pharmacology , Wakefulness-Promoting Agents/therapeutic use , Young Adult
17.
Front Biosci (Landmark Ed) ; 24(3): 564-575, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30468674

ABSTRACT

Modafinil (Mo) is increasingly being used as an enhancement drug rather than for its therapeutic effects. The effects of this drug have been examined in attention deficit disorders, depression, mental fatigue, and in enhancing concentration. The drug possesses wakefulness-promoting properties which are mediated through the interaction of orexinergic system with the activated sympathetic nervous system. Mo exerts a synergistic effect on the orexin system, controls energy expenditure and strengthens the ability of the individual to exercise. Some view Mo as a drug that enhances sports performance, since it induces a prolonged wakefulness and decreasing the sense of fatigue. These characteristics being similar to conventional stimulants have allowed Mo to emerge as a novel stimulant requiring medico-legal considerations. However, more studies are needed to better understand the mid and long-term effects of the drug on user/abuser.


Subject(s)
Central Nervous System/drug effects , Modafinil/pharmacology , Orexins/metabolism , Wakefulness/drug effects , Animals , Attention/drug effects , Attention/physiology , Central Nervous System/metabolism , Central Nervous System/physiology , Cognition/drug effects , Cognition/physiology , Humans , Modafinil/metabolism , Modafinil/pharmacokinetics , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Wakefulness/physiology , Wakefulness-Promoting Agents/metabolism , Wakefulness-Promoting Agents/pharmacokinetics , Wakefulness-Promoting Agents/pharmacology
18.
Neurochem Res ; 43(8): 1511-1518, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29876791

ABSTRACT

Cannabis sativa is a plant that contains more than 500 components, of which the most studied are Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD). Several studies have indicated that CBD displays neurobiological effects, including wake promotion. Moreover, experimental evidence has shown that injections of CBD enhance wake-related compounds, such as monoamines (dopamine, serotonin, epinephrine, and norepinephrine). However, no clear evidence is available regarding the effects of CBD on additional wake-related neurochemicals such as acetylcholine (ACh). Here, we demonstrate that systemic injections of CBD (0, 5, 10 or 30 mg/kg, i.p.) at the beginning of the lights-on period, increase the extracellular levels of ACh collected from the basal forebrain and measured by microdialysis and HPLC means. Moreover, the time course effects on the contents of ACh were present 5 h post-injection of CBD. Altogether, these data demonstrate that CBD increases ACh levels in a brain region related to wake control. This study is the first to show the effects of ACh levels in CBD-treated rats and suggests that the basal forebrain might be a site of action of CBD for wakefulness modulation.


Subject(s)
Acetylcholine/metabolism , Basal Forebrain/drug effects , Cannabidiol/pharmacology , Animals , Cannabidiol/administration & dosage , Male , Rats, Wistar , Time Factors , Wakefulness/drug effects , Wakefulness-Promoting Agents/administration & dosage , Wakefulness-Promoting Agents/pharmacology
19.
Int J Neuropsychopharmacol ; 21(7): 687-696, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29635319

ABSTRACT

Background: Modafinil, a nonamphetaminic wake-promoting compound, is prescribed as first line therapy in narcolepsy, an invalidating disorder characterized by excessive daytime sleepiness and cataplexy. Although its mode of action remains incompletely known, recent studies indicated that modafinil modulates astroglial connexin-based gap junctional communication as administration of a low dose of flecainide, an astroglial connexin inhibitor, enhanced the wake-promoting and procognitive activity of modafinil in rodents and healthy volunteers. The aim of this study is to investigate changes in glucose cerebral metabolism in rodents, induced by the combination of modafinil+flecainide low dose (called THN102). Methods: The impact of THN102 on brain glucose metabolism was noninvasively investigated using 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography imaging in Sprague-Dawley male rats. Animals were injected with vehicle, flecainide, modafinil, or THN102 and further injected with 18F-2-fluoro-2-deoxy-D-glucose followed by 60-minute Positron Emission Tomography acquisition. 18F-2-fluoro-2-deoxy-D-glucose Positron Emission Tomography images were coregistered to a rat brain template and normalized from the total brain Positron Emission Tomography signal. Voxel-to-voxel analysis was performed using SPM8 software. Comparison of brain glucose metabolism between groups was then performed. Results: THN102 significantly increased regional brain glucose metabolism as it resulted in large clusters of 18F-2-fluoro-2-deoxy-D-glucose uptake localized in the cortex, striatum, and amygdala compared with control or drugs administered alone. These regions, highly involved in the regulation of sleep-wake cycle, emotions, and cognitive functions were hence quantitatively modulated by THN102. Conclusion: Data presented here provide the first evidence of a regional brain activation induced by THN102, currently being tested in a phase II clinical trial in narcoleptic patients.


Subject(s)
Amygdala/drug effects , Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Flecainide/pharmacology , Fluorodeoxyglucose F18/pharmacokinetics , Modafinil/pharmacology , Positron-Emission Tomography/methods , Voltage-Gated Sodium Channel Blockers/pharmacology , Wakefulness-Promoting Agents/pharmacology , Amygdala/diagnostic imaging , Amygdala/metabolism , Animals , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Corpus Striatum/diagnostic imaging , Corpus Striatum/metabolism , Drug Combinations , Flecainide/administration & dosage , Male , Modafinil/administration & dosage , Rats , Rats, Sprague-Dawley , Voltage-Gated Sodium Channel Blockers/administration & dosage , Wakefulness-Promoting Agents/administration & dosage
20.
Article in English | MEDLINE | ID: mdl-28499899

ABSTRACT

The wake-promoting drug Modafinil has been used for many years for treatment of Narcolepsy and Excessive Daytime Sleepiness, due to a dopamine-related psychostimulant action. Recent studies have indicated that Modafinil prevents neuroinflammation in animal models. Thus, the aim of the present study was to evaluate the effect of Modafinil pretreatment in the Lipopolysaccharide (LPS)-induced sickness and depressive-like behaviors. Adult male C57BL/6J mice were pretreated with Vehicle or Modafinil (90mg/Kg) and, 30min later, received a single saline or LPS (2mg/Kg) administration, and were submitted to the open field and elevated plus maze test 2h later. After 24h, mice were subjected to tail suspension test, followed by either flow cytometry with whole brain for CD11b+CD45+ cells or qPCR in brain areas for cytokine gene expression. Modafinil treatment prevented the LPS-induced motor impairment, anxiety-like and depressive-like behaviors, as well as the increase in brain CD11b+CD45high cells induced by LPS. Our results indicate that Modafinil pretreatment also decreased the IL-1ß gene upregulation caused by LPS in brain areas, which is possibly correlated with the preventive behavioral effects. The pharmacological blockage of the dopaminergic D1R by the drug SCH-23390 counteracted the effect of Modafinil on locomotion and anxiety-like behavior, but not on depressive-like behavior and brain immune cells. The dopaminergic D1 receptor signaling is essential to the Modafinil effects on LPS-induced alterations in locomotion and anxiety, but not on depression and brain macrophages. This evidence suggests that Modafinil treatment might be useful to prevent inflammation-related behavioral alterations, possibly due to a neuroimmune mechanism.


Subject(s)
Benzhydryl Compounds/pharmacology , Dopamine Agents/pharmacology , Illness Behavior/drug effects , Motor Activity/drug effects , Movement Disorders/drug therapy , Receptors, Dopamine D1/metabolism , Animals , Anxiety/drug therapy , Anxiety/metabolism , Anxiety/pathology , Benzazepines/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Movement/drug effects , Cell Movement/physiology , Depression/drug therapy , Depression/metabolism , Depression/pathology , Disease Models, Animal , Escherichia coli , Illness Behavior/physiology , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Modafinil , Motor Activity/physiology , Movement Disorders/etiology , Movement Disorders/metabolism , Movement Disorders/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Receptors, Dopamine D1/antagonists & inhibitors , Wakefulness-Promoting Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...