Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Endocrinology ; 165(3)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38279940

ABSTRACT

The arcuate nucleus kisspeptin (ARNKISS) neurons represent the GnRH pulse generator that likely drives pulsatile gonadotropin secretion in all mammals. Using an improved GCaMP fiber photometry system enabling long-term continuous recordings, we aimed to establish a definitive profile of ARNKISS neuronal activity across the murine estrous cycle. As noted previously, a substantial reduction in the frequency of ARNKISS neuron synchronization events (SEs) occurs on late proestrus and extends into estrus. The SE amplitude remains constant throughout the cycle. During metestrus, we unexpectedly detected many multipeak SEs where many SEs occurred rapidly, within 160 seconds of each other. By applying a machine learning-based, k-means clustering analysis, we were further able to detect substantial within-stage variability in the patterns of pulse generator activity. Estrous cycle-dependent changes in SE activity occurred around the time of lights on and off. We also find that a mild stressor such as vaginal lavage reduces ARNKISS neuron SE frequency for up to 3 hours. These observations provide a comprehensive account of ARNKISS neuron activity across the estrous cycle, highlight a new pattern of multipeak SE activity, and introduce a new k-means clustering approach for analyzing ARNKISS neuron population behavior.


Subject(s)
Gonadotropin-Releasing Hormone , Luteinizing Hormone , Animals , Female , Mice , Arcuate Nucleus of Hypothalamus/metabolism , Estrous Cycle/physiology , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Neurons/metabolism
2.
J Neurosci ; 43(5): 846-862, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36564184

ABSTRACT

Stress disorders impair sleep and quality of life; however, their pathomechanisms are unknown. Prolactin-releasing peptide (PrRP) is a stress mediator; we therefore hypothesized that PrRP may be involved in the development of stress disorders. PrRP is produced by the medullary A1/A2 noradrenaline (NA) cells, which transmit stress signals to forebrain centers, and by non-NA cells in the hypothalamic dorsomedial nucleus. We found in male rats that both PrRP and PrRP-NA cells innervate melanin-concentrating hormone (MCH) producing neurons in the dorsolateral hypothalamus (DLH). These cells serve as a key hub for regulating sleep and affective states. Ex vivo, PrRP hyperpolarized MCH neurons and further increased the hyperpolarization caused by NA. Following sleep deprivation, intracerebroventricular PrRP injection reduced the number of REM sleep-active MCH cells. PrRP expression in the dorsomedial nucleus was upregulated by sleep deprivation, while downregulated by REM sleep rebound. Both in learned helplessness paradigm and after peripheral inflammation, impaired coping with sustained stress was associated with (1) overactivation of PrRP cells, (2) PrRP protein and receptor depletion in the DLH, and (3) dysregulation of MCH expression. Exposure to stress in the PrRP-insensitive period led to increased passive coping with stress. Normal PrRP signaling, therefore, seems to protect animals against stress-related disorders. PrRP signaling in the DLH is an important component of the PrRP's action, which may be mediated by MCH neurons. Moreover, PrRP receptors were downregulated in the DLH of human suicidal victims. As stress-related mental disorders are the leading cause of suicide, our findings may have particular translational relevance.SIGNIFICANCE STATEMENT Treatment resistance to monoaminergic antidepressants is a major problem. Neuropeptides that modulate the central monoaminergic signaling are promising targets for developing alternative therapeutic strategies. We found that stress-responsive prolactin-releasing peptide (PrRP) cells innervated melanin-concentrating hormone (MCH) neurons that are crucial in the regulation of sleep and mood. PrRP inhibited MCH cell activity and enhanced the inhibitory effect evoked by noradrenaline, a classic monoamine, on MCH neurons. We observed that impaired PrRP signaling led to failure in coping with chronic/repeated stress and was associated with altered MCH expression. We found alterations of the PrRP system also in suicidal human subjects. PrRP dysfunction may underlie stress disorders, and fine-tuning MCH activity by PrRP may be an important part of the mechanism.


Subject(s)
Hypothalamic Hormones , Sleep Deprivation , Rats , Male , Humans , Animals , Prolactin-Releasing Hormone/pharmacology , Prolactin-Releasing Hormone/metabolism , Sleep Deprivation/metabolism , Mood Disorders/etiology , Quality of Life , Rats, Wistar , Hypothalamic Hormones/metabolism , Sleep/physiology , Neurons/physiology , Norepinephrine/metabolism
3.
PLoS One ; 16(5): e0251767, 2021.
Article in English | MEDLINE | ID: mdl-33984047

ABSTRACT

Sleep disturbance is a common and disruptive symptom of neurodegenerative diseases such as Alzheimer's and Huntington's disease (HD). In HD patients, sleep fragmentation appears at an early stage of disease, although features of the earliest sleep abnormalities in presymptomatic HD are not fully established. Here we used novel automated analysis of quantitative electroencephalography to study transitions between wake and non-rapid eye movement sleep in a sheep model of presymptomatic HD. We found that while the number of transitions between sleep and wake were similar in normal and HD sheep, the dynamics of transitions from sleep-to-wake differed markedly between genotypes. Rather than the gradual changes in EEG power that occurs during transitioning from sleep-to-wake in normal sheep, transition into wake was abrupt in HD sheep. Furthermore, transitions to wake in normal sheep were preceded by a significant reduction in slow wave power, whereas in HD sheep this prior reduction in slow wave power was far less pronounced. This suggests an impaired ability to prepare for waking in HD sheep. The abruptness of awakenings may also have potential to disrupt sleep-dependent processes if they are interrupted in an untimely and disjointed manner. We propose that not only could these abnormal dynamics of sleep transitions be useful as an early biomarker of HD, but also that our novel methodology would be useful for studying transition dynamics in other sleep disorders.


Subject(s)
Huntington Disease/complications , Sleep Deprivation/physiopathology , Animals , Disease Models, Animal , Electroencephalography , Female , Humans , Huntington Disease/physiopathology , Polysomnography/methods , Sheep, Domestic , Sleep/physiology , Sleep Deprivation/diagnosis , Sleep Deprivation/etiology , Wakefulness/physiology
4.
Neurobiol Dis ; 155: 105367, 2021 07.
Article in English | MEDLINE | ID: mdl-33848636

ABSTRACT

Sleep disruption is a common invisible symptom of neurological dysfunction in Huntington's disease (HD) that takes an insidious toll on well-being of patients. Here we used electroencephalography (EEG) to examine sleep in 6 year old OVT73 transgenic sheep (Ovis aries) that we used as a presymptomatic model of HD. We hypothesized that despite the lack of overt symptoms of HD at this age, early alterations of the sleep-wake pattern and EEG powers may already be present. We recorded EEG from female transgenic and normal sheep (5/group) during two undisturbed 'baseline' nights with different lighting conditions. We then recorded continuously through a night of sleep disruption and the following 24 h (recovery day and night). On baseline nights, regardless of whether the lights were on or off, transgenic sheep spent more time awake than normal sheep particularly at the beginning of the night. Furthermore, there were significant differences between transgenic and normal sheep in both EEG power and its pattern of distribution during non-rapid eye movement (NREM) sleep. In particular, there was a significant decrease in delta (0.5-4 Hz) power across the night in transgenic compared to normal sheep, and the distributions of delta, theta and alpha oscillations that typically dominate the EEG in the first half of the night of normal sheep were skewed so they were predominant in the second, rather than the first half of the night in transgenic sheep. Interestingly, the effect of sleep disruption on normal sheep was also to skew the pattern of distribution of EEG powers so they looked more like that of transgenic sheep under baseline conditions. Thus it is possible that transgenic sheep exist in a state that resemble a chronic state of physiological sleep deprivation. During the sleep recovery period, normal sheep showed a significant 'rebound' increase in delta power with frontal dominance. A similar rebound was not seen in transgenic sheep, suggesting that their homeostatic response to sleep deprivation is abnormal. Although sleep abnormalities in early stage HD patients are subtle, with patients often unaware of their existence, they may contribute to impairment of neurological function that herald the onset of disease. A better understanding of the mechanisms underlying EEG abnormalities in early stage HD would give insight into how, and when, they progress into the sleep disorder. The transgenic sheep model is ideally positioned for studies of the earliest phase of disease when sleep abnormalities first emerge.


Subject(s)
Electroencephalography/methods , Huntington Disease/genetics , Huntington Disease/physiopathology , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , Sleep Stages/physiology , Animals , Animals, Genetically Modified , Disease Models, Animal , Female , Sheep
5.
Neurotherapeutics ; 17(3): 1075-1086, 2020 07.
Article in English | MEDLINE | ID: mdl-32297185

ABSTRACT

Huntington's disease (HD) is characterised by progressive symptoms including cognitive deficits and sleep/wake disturbances reflected in an abnormal electroencephalography (EEG). Modafinil, a wake-promoting and cognitive-enhancing drug, has been considered as a treatment for HD. We used HD (R6/2) mice to investigate the potential for using modafinil to treat sleep-wake disturbance in HD. R6/2 mice show sleep-wake and EEG changes similar to those seen in HD patients, with increased rapid eye movement sleep (REMS), decreased wakefulness/increased non-REMS (NREMS), and pathological changes in EEG spectra, particularly an increase in gamma power. We recorded EEG from R6/2 and wild-type mice treated with modafinil acutely (with single doses between 25 and 100 mg/kg; at 12 and 16 weeks of age), or chronically (64 mg/kg modafinil/day from 6 to 15 weeks). Acutely, modafinil increased wakefulness in R6/2 mice and restored NREMS to wild-type levels at 12 weeks. It also suppressed the pathologically increased REMS. This was accompanied by decreased delta power, increased peak frequency of theta, and increased gamma power. At 16 weeks, acute modafinil also restored wakefulness and NREMS to wild-type levels. However, whilst REMS decreased, it did not return to normal levels. By contrast, in the chronic treatment group, modafinil-induced wakefulness was maintained at 15 weeks (after 9 weeks of treatment). Interestingly, chronic modafinil also caused widespread suppression of power across the EEG spectra, including a reduction in gamma that increases pathologically in R6/2 mice. The complex EEG effects of modafinil in R6/2 mice should provide a baseline for further studies to investigate the translatability of these result to clinical practice.


Subject(s)
Electroencephalography/methods , Huntington Disease/drug therapy , Modafinil/administration & dosage , Wakefulness-Promoting Agents/administration & dosage , Wakefulness/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Electroencephalography/drug effects , Huntington Disease/genetics , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sleep Stages/drug effects , Sleep Stages/physiology , Wakefulness/physiology
6.
eNeuro ; 7(2)2020.
Article in English | MEDLINE | ID: mdl-32122958

ABSTRACT

Sleep spindles are distinctive transient patterns of brain activity that typically occur during non-rapid eye movement (NREM) sleep in humans and other mammals. Thought to be important for the consolidation of learning, they may also be useful for indicating the progression of aging and neurodegenerative diseases. The aim of this study was to characterize sleep spindles in sheep (Ovis aries). We recorded electroencephalographs wirelessly from six sheep over a continuous period containing 2 nights and a day. We detected and characterized spindles using an automated algorithm. We found that sheep sleep spindles fell within the classical range seen in humans (10-16 Hz), but we did not see a further separation into fast and slow bands. Spindles were detected predominantly during NREM sleep. Spindle characteristics (frequency, duration, density, topography) varied between individuals, but were similar within individuals between nights. Spindles that occurred during NREM sleep in daytime were indistinguishable from those found during NREM sleep at night. Surprisingly, we also detected numerous spindle-like events during unequivocal periods of wake during the day. These events were mainly local (detected at single sites), and their characteristics differed from spindles detected during sleep. These "wake spindles" are likely to be events that are commonly categorized as "spontaneous alpha activity" during wake. We speculate that wake and sleep spindles are generated via different mechanisms, and that wake spindles play a role in cognitive processes that occur during the daytime.


Subject(s)
Sleep, Slow-Wave , Sleep , Animals , Electroencephalography , Learning , Polysomnography , Sheep
7.
Front Pharmacol ; 10: 831, 2019.
Article in English | MEDLINE | ID: mdl-31404291

ABSTRACT

Study Objectives: (a) To describe the microarchitecture of wakefulness and sleep following administrations of 5- and 10-mg/kg AM-251 in rats. (b) To develop a new statistical method to follow bout-to-bout dynamics. Method: Wistar rats (n = 6) had been equipped with electroencephalography (EEG) and electromyography (EMG) electrodes. Following their recovery and habituation after the surgery, the animals were injected with vehicle and 5- and 10-mg/kg AM-251 intraperitoneally and EEG, EMG, and motor activity were analyzed for the subsequent 3 h. Results: AM-251 induced a dose- and time-dependent increase in the number of bouts in active wake (AW), and it decreased this number in all other vigilance states except in passive wake (PW). In contrast, the bout duration in PW compensatory decreased. The effect of AM-251 on the sleep transition dynamics was monitored with a new tool we call "transition heatmap." The analysis of bout trajectories with transition heatmaps reveals a highly organized pattern. Conclusion: AM-251 selectively influences the frequency of vigilance state transitions, but it has no direct impact on the state lengths. AM-251 markedly changed the state transition dynamics, which was visualized with the help of state transition heatmaps.

8.
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
9.
Pharmacology ; 103(3-4): 151-158, 2019.
Article in English | MEDLINE | ID: mdl-30673678

ABSTRACT

The endocannabinoid and serotonin (5-HT) systems have key roles in the regulation of several physiological functions such as motor activity and food intake but also in the development of psychiatric disorders. Here we tested the hypothesis, whether blockade of serotonin 2C (5-HT2C) receptors prevents the reduced locomotor activity and other behavioral effects caused by a cannabinoid 1 (CB1) receptor antagonist. As a pretreatment, we administered SB-242084 (1 mg/kg, ip.), a 5-HT2C receptor antagonist or vehicle (VEH) followed by the treatment with AM-251 (5 or 10 mg/kg, ip.), a CB1 receptor antagonist or VEH. The effects of the two drugs alone or in co-administration were investigated in social interaction (SI) and elevated plus maze (EPM) tests in male Wistar rats. Our results show that AM-251 decreased the time spent with rearing in the SI test and decreased locomotor activity in EPM test. In contrast, SB-242084 produced increased locomotor activity in SI test and evoked anxiolytic-like effect in both SI and EPM tests. When applied the drugs in combination, these behavioral effects of AM-251 were moderated by SB-242084. Based on these findings, we conclude that certain unwanted behavioral effects of CB1 receptor antagonists could be prevented by pretreatment with 5-HT2C receptor antagonists.


Subject(s)
Aminopyridines/pharmacology , Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Indoles/pharmacology , Locomotion/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Serotonin, 5-HT2C/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Cannabinoid Receptor Antagonists/toxicity , Exploratory Behavior/drug effects , Male , Maze Learning/drug effects , Piperidines/toxicity , Pyrazoles/toxicity , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Social Behavior
11.
Front Pharmacol ; 10: 1636, 2019.
Article in English | MEDLINE | ID: mdl-32063851

ABSTRACT

Serotonin 2C receptors (5-HT2CRs) are implicated in the pathomechanism and treatment of anxiety and depression. Recently, as a new biomarker of depression, alterations in the gamma power of the electroencephalogram (EEG) have been suggested. Chronic treatment with the selective serotonin reuptake inhibitor (SSRI) antidepressant escitalopram has been shown to cause sleep-wake stage-dependent alterations in gamma power. However, despite the antidepressant potency of 5-HT2CR-antagonists, there is no data available regarding the effects of selective 5-HT2CR-antagonists on gamma activity. Therefore, we investigate the acute effect of the 5-HT2CR-antagonist SB-242084 on gamma power in different vigilance stages when given in monotherapy, or in combination with chronic escitalopram treatment. We administered SB-242084 (1 mg/kg, intraperitoneally) or vehicle to EEG-equipped rats after a 21-day-long pretreatment with escitalopram (10 mg/kg/day, via osmotic minipumps) or vehicle. Frontoparietal EEG, electromyogram, and motor activity were recorded during the first 3 h of passive phase, after the administration of SB-242084. Quantitative EEG analysis revealed that acute SB-242084 increased gamma power (30-60 Hz) in light and deep slow-wave sleep, and passive wakefulness. However, in active wakefulness, rapid eye movement sleep, and intermediate stage, no change was observed in gamma power. The profile of the effect of SB-242084 on gamma power was similar to that produced by chronic escitalopram. Moreover, SB-242084 did not alter chronic escitalopram-induced effects on gamma. In conclusion, the similarity in the effect of the 5-HT2CR-antagonist and chronic SSRI on gamma power provides further evidence for the therapeutic potential of 5-HT2CR-antagonists in the treatment of depression and/or anxiety.

12.
Eur J Pharm Sci ; 121: 347-355, 2018 08 30.
Article in English | MEDLINE | ID: mdl-29908300

ABSTRACT

Brain oscillations in the gamma frequency band of the electroencephalogram (EEG) have been implicated in several sensory and cognitive processes, and have also been associated with numerous neuropsychiatric disorders, including depression. The widely prescribed selective serotonin reuptake inhibitors (SSRIs), similarly to other antidepressants, are known to produce markedly different effects on sleep and behavioral measures with acute and chronic administration. Although there are studies examining the acute effect of escitalopram on slower (<30 Hz) oscillations, we hardly could find any data about the effect of the drug on higher-frequency EEG oscillations (>30 Hz) in different sleep-wake stages, particularly comparing the acute and chronic effects of the drug concerning gamma oscillations. Our aim was to investigate, how escitalopram affects gamma power in different sleep-wake stages, and to discover possible differential effects between acute and chronic treatment. EEG-equipped Wistar rats were treated with escitalopram or vehicle acutely (10 mg/kg, i.p.) or chronically (10 mg/kg/day for 21 days, osmotic minipumps) and frontoparietal EEG, electromyogram and motor activity were recorded during the first 3 h of passive phase. We found that acute and chronic escitalopram treatment affected gamma oscillations differently. While acute escitalopram caused a reduction in gamma power during rapid eye movement sleep (REMS) and intermediate stage of sleep (IS), chronic treatment caused an elevation in gamma power during non-REMS stages, namely in light and deep slow-wave sleep (SWS-1 and SWS-2, respectively) and in IS. However, gamma activity during active and passive wakefulness (AW and PW, respectively) was not influenced by either acute or chronic dosing of escitalopram. Furthermore, we found that in drug-free (vehicle-treated) rats, a relatively high gamma power was present during wakefulness and REMS, while a much lower power was measured during non-REMS stages. These findings indicate that acute and chronic administration of escitalopram alter gamma activity differently, moreover, in a sleep-wake stage dependent manner that may be related to differential therapeutic and/or side effects.


Subject(s)
Antidepressive Agents/administration & dosage , Citalopram/administration & dosage , Electroencephalography/drug effects , Sleep Stages/drug effects , Animals , Drug Administration Schedule , Electromyography , Male , Rats, Wistar
13.
Brain Behav Immun ; 56: 96-104, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26891860

ABSTRACT

Interleukin-1ß is one of the main mediators in the cross-talk between the immune system and the central nervous system. Higher interleukin-1ß levels are found in mood spectrum disorders, and the stress-induced expression rate of the interleukin-1ß gene (IL1B) is altered by polymorphisms in the region. Therefore we examined the effects of rs16944 and rs1143643 single nucleotide polymorphisms (SNPs) within the IL1B gene on depressive and anxiety symptoms, as measured by the Brief Symptom Inventory, in a Hungarian population sample of 1053 persons. Distal and proximal environmental stress factors were also included in our analysis, namely childhood adversity and recent negative life-events. We found that rs16944 minor (A) allele specifically interacted with childhood adversity increasing depressive and anxiety symptoms, while rs1143643's minor (A) allele showed protective effect against depressive symptoms after recent life stress. The genetic main effects of the two SNPs were not significant in the main analysis, but the interaction effects remained significant after correction for multiple testing. In addition, the effect of rs16944 A allele was reversed in a subsample with low-exposure to life stress, suggesting a protective effect against depressive symptoms, in the post hoc analysis. In summary, both of the two IL1B SNPs showed specific environmental stressor-dependent effects on mood disorder symptoms. We also demonstrated that the presence of exposure to childhood adversity changed the direction of the rs16944 effect on depression phenotype. Therefore our results suggest that it is advisable to include environmental factors in genetic association studies when examining the effect of the IL1B gene.


Subject(s)
Adult Survivors of Child Adverse Events/statistics & numerical data , Anxiety Disorders , Depressive Disorder , Gene-Environment Interaction , Interleukin-1beta/genetics , Stress, Psychological , Adolescent , Adult , Anxiety Disorders/epidemiology , Anxiety Disorders/etiology , Anxiety Disorders/genetics , Depressive Disorder/epidemiology , Depressive Disorder/etiology , Depressive Disorder/genetics , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Stress, Psychological/epidemiology , Young Adult
14.
J Neural Transm (Vienna) ; 123(5): 541-8, 2016 05.
Article in English | MEDLINE | ID: mdl-26821321

ABSTRACT

Interleukin-6 (IL-6) has emerged as a potent biomarker for depression as its elevated plasma levels in patients with clinical depression have been confirmed by meta-analyses. Increased plasma IL-6 concentration was associated with various psychological stress factors and physical disorders accompanied by pain. Another modulator of the IL-6 level is rs1800795, a promoter polymorphism in the IL-6 gene which is able to influence its expression rate. Therefore, we examined in a Hungarian population sample of 1053 volunteers with European origins if rs1800795 polymorphism can affect depression symptoms measured by Zung Self-rating Depression Scale (ZSDS), and Brief Symptom Inventory (BSI). We also investigated the interactions of the polymorphism with reported painful physical conditions and Recent Negative Life Events (RLE) measured by the List of Life Threatening Experiences. Rs1800795 significantly interacted with both RLE and painful condition on depressive symptoms measured by ZSDS and BSI using different heritability models, while no main effects of the polymorphism were identified. After correction for multiple testing only the rs1800795 × RLE interaction effect (recessive model) remained significant on the BSI score, while both RLE and painful conditions significantly interacted on the ZSDS. In conclusion, the functional IL-6 rs1800795 polymorphism in interaction with various stress factors increases the risk of depression and has a greater impact on symptoms measured by the ZSDS. Thus, IL-6 and other cytokines may be more relevant in the development of somatic symptoms compared to affective signs of depression, delineating a specific genotype-phenotype relationship in this heterogeneous disorder.


Subject(s)
Depressive Disorder/etiology , Interleukin-6/genetics , Pain/complications , Pain/genetics , Polymorphism, Single Nucleotide/genetics , Stress, Psychological/complications , Stress, Psychological/genetics , Adolescent , Adult , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Promoter Regions, Genetic/genetics , Psychiatric Status Rating Scales , Young Adult
15.
Brain Struct Funct ; 221(7): 3521-46, 2016 09.
Article in English | MEDLINE | ID: mdl-26462664

ABSTRACT

Neuropeptide S (NPS) is a regulatory peptide expressed by limited number of neurons in the brainstem. The simultaneous anxiolytic and arousal-promoting effect of NPS suggests an involvement in mood control and vigilance, making the NPS-NPS receptor system an interesting potential drug target. Here we examined, in detail, the distribution of NPS-immunoreactive (IR) fiber arborizations in brain regions of rat known to be involved in the regulation of sleep and arousal. Such nerve terminals were frequently apposed to GABAergic/galaninergic neurons in the ventro-lateral preoptic area (VLPO) and to tyrosine hydroxylase-IR neurons in all hypothalamic/thalamic dopamine cell groups. Then we applied the single platform-on-water (mainly REM) sleep deprivation method to study the functional role of NPS in the regulation of arousal. Of the three pontine NPS cell clusters, the NPS transcript levels were increased only in the peri-coerulear group in sleep-deprived animals, but not in stress controls. The density of NPS-IR fibers was significantly decreased in the median preoptic nucleus-VLPO region after the sleep deprivation, while radioimmunoassay and mass spectrometry measurements showed a parallel increase of NPS in the anterior hypothalamus. The expression of the NPS receptor was, however, not altered in the VLPO-region. The present results suggest a selective activation of one of the three NPS-expressing neuron clusters as well as release of NPS in distinct forebrain regions after sleep deprivation. Taken together, our results emphasize a role of the peri-coerulear cluster in the modulation of arousal, and the importance of preoptic area for the action of NPS on arousal and sleep.


Subject(s)
Arousal , Brain/cytology , Brain/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Animals , Anterior Hypothalamic Nucleus/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Galanin/metabolism , Glutamic Acid/metabolism , Locus Coeruleus/metabolism , Male , Preoptic Area/cytology , Preoptic Area/metabolism , RNA, Messenger , Rats , Rats, Wistar , Receptors, Neuropeptide/metabolism , Sleep , Sleep Deprivation/metabolism
16.
BMC Neurosci ; 15: 120, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406958

ABSTRACT

BACKGROUND: Shortened rapid eye movement (REM) sleep latency and increased REM sleep amount are presumed biological markers of depression. These sleep alterations are also observable in several animal models of depression as well as during the rebound sleep after selective REM sleep deprivation (RD). Furthermore, REM sleep fragmentation is typically associated with stress procedures and anxiety. The selective serotonin reuptake inhibitor (SSRI) antidepressants reduce REM sleep time and increase REM latency after acute dosing in normal condition and even during REM rebound following RD. However, their therapeutic outcome evolves only after weeks of treatment, and the effects of chronic treatment in REM-deprived animals have not been studied yet. RESULTS: Chronic escitalopram- (10 mg/kg/day, osmotic minipump for 24 days) or vehicle-treated rats were subjected to a 3-day-long RD on day 21 using the flower pot procedure or kept in home cage. On day 24, fronto-parietal electroencephalogram, electromyogram and motility were recorded in the first 2 h of the passive phase. The observed sleep patterns were characterized applying standard sleep metrics, by modelling the transitions between sleep phases using Markov chains and by spectral analysis. Based on Markov chain analysis, chronic escitalopram treatment attenuated the REM sleep fragmentation [accelerated transition rates between REM and non-REM (NREM) stages, decreased REM sleep residence time between two transitions] during the rebound sleep. Additionally, the antidepressant avoided the frequent awakenings during the first 30 min of recovery period. The spectral analysis showed that the SSRI prevented the RD-caused elevation in theta (5-9 Hz) power during slow-wave sleep. Conversely, based on the aggregate sleep metrics, escitalopram had only moderate effects and it did not significantly attenuate the REM rebound after RD. CONCLUSION: In conclusion, chronic SSRI treatment is capable of reducing several effects on sleep which might be the consequence of the sub-chronic stress caused by the flower pot method. These data might support the antidepressant activity of SSRIs, and may allude that investigating the rebound period following the flower pot protocol could be useful to detect antidepressant drug response. Markov analysis is a suitable method to study the sleep pattern.


Subject(s)
Brain/drug effects , Citalopram/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sleep Deprivation/physiopathology , Sleep, REM/drug effects , Animals , Brain/physiopathology , Catheters, Indwelling , Electrodes, Implanted , Electroencephalography , Male , Markov Chains , Models, Neurological , Polysomnography , Random Allocation , Rats, Wistar , Sleep, REM/physiology , Theta Rhythm/drug effects
17.
Proc Natl Acad Sci U S A ; 111(35): E3735-44, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136085

ABSTRACT

Narcolepsy is a chronic sleep disorder, likely with an autoimmune component. During 2009 and 2010, a link between A(H1N1)pdm09 Pandemrix vaccination and onset of narcolepsy was suggested in Scandinavia. In this study, we searched for autoantibodies related to narcolepsy using a neuroanatomical array: rat brain sections were processed for immunohistochemistry/double labeling using patient sera/cerebrospinal fluid as primary antibodies. Sera from 89 narcoleptic patients, 52 patients with other sleep-related disorders (OSRDs), and 137 healthy controls were examined. Three distinct patterns of immunoreactivity were of particular interest: pattern A, hypothalamic melanin-concentrating hormone and proopiomelanocortin but not hypocretin/orexin neurons; pattern B, GABAergic cortical interneurons; and pattern C, mainly globus pallidus neurons. Altogether, 24 of 89 (27%) narcoleptics exhibited pattern A or B or C. None of the patterns were exclusive for narcolepsy but were also detected in the OSRD group at significantly lower numbers. Also, some healthy controls exhibited these patterns. The antigen of pattern A autoantibodies was identified as the common C-terminal epitope of neuropeptide glutamic acid-isoleucine/α-melanocyte-stimulating hormone (NEI/αMSH) peptides. Passive transfer experiments on rat showed significant effects of pattern A human IgGs on rapid eye movement and slow-wave sleep time parameters in the inactive phase and EEG θ-power in the active phase. We suggest that NEI/αMSH autoantibodies may interfere with the fine regulation of sleep, contributing to the complex pathogenesis of narcolepsy and OSRDs. Also, patterns B and C are potentially interesting, because recent data suggest a relevance of those brain regions/neuron populations in the regulation of sleep/arousal.


Subject(s)
Autoantibodies/blood , Brain/immunology , Brain/pathology , Narcolepsy/immunology , Narcolepsy/pathology , Sleep/physiology , Adolescent , Adult , Animals , Autoantibodies/immunology , Colchicine/analogs & derivatives , Colchicine/pharmacology , Electroencephalography , Globus Pallidus/immunology , Globus Pallidus/pathology , Hippocampus/immunology , Hippocampus/pathology , Humans , Immunoglobulin G/blood , Interneurons/immunology , Interneurons/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/immunology , Neocortex/pathology , Nerve Tissue Proteins/metabolism , Olfactory Bulb/immunology , Olfactory Bulb/pathology , Rats , Rats, Wistar , Young Adult
18.
Exp Brain Res ; 232(3): 935-46, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24395141

ABSTRACT

Several multi-target drugs used in treating psychiatric disorders, such as antidepressants (e.g. agomelatine, trazodone, nefazodone, amitriptyline, mirtazapine, mianserin, fluoxetine) or most atypical antipsychotics, have 5-hydroxytryptamine 2C (5-HT2C) receptor-blocking property. Adaptive changes in 5-HT2C receptor-mediated functions are suggested to contribute to therapeutic effects of selective serotonin reuptake inhibitor (SSRI) antidepressants after weeks of treatment, at least in part. Beyond the mediation of anxiety and other functions, 5-HT2C receptors are involved in sleep regulation. Anxiety-related adaptive changes caused by antidepressants have been studied extensively, although sleep- and electroencephalography (EEG)-related functional studies are still lacking. The aim of this study was to investigate the effects of chronic SSRI treatment on 5-HT2C receptor antagonist-induced functions in different vigilance stages and on quantitative EEG (Q-EEG) spectra. Rats were treated with a single dose of the selective 5-HT2C receptor antagonist SB-242084 (1 mg/kg, i.p.) or vehicle at the beginning of passive phase following a 20-day-long SSRI (escitalopram; 10 mg/kg/day, osmotic minipump) or VEHICLE pretreatment. Fronto-parietal electroencephalogram, electromyogram and motility were recorded during the first 3 h of passive phase. We found that the chronic escitalopram pretreatment attenuated the SB-242084-caused suppression in rapid eye movement sleep (REMS). On the contrary, the 5-HT2C receptor antagonist-induced elevations in passive wake and theta (5-9 Hz) power density during active wake and REMS were not affected by the SSRI. In conclusion, attenuation in certain, but not all vigilance- and Q-EEG-related functions induced by the 5-HT2C receptor antagonist, suggests dissociation in 5-HT2C receptor adaptation.


Subject(s)
Adaptation, Physiological/drug effects , Aminopyridines/pharmacology , Citalopram/pharmacology , Indoles/pharmacology , Serotonin Antagonists/pharmacology , Sleep, REM/drug effects , Theta Rhythm/drug effects , Wakefulness/drug effects , Analysis of Variance , Animals , Electroencephalography , Electromyography , Fourier Analysis , Male , Rats , Rats, Wistar , Reaction Time/drug effects , Selective Serotonin Reuptake Inhibitors
19.
PLoS One ; 8(4): e59809, 2013.
Article in English | MEDLINE | ID: mdl-23560056

ABSTRACT

STUDY OBJECTIVES: Millions suffer from sleep disorders that often accompany severe illnesses such as major depression; a leading psychiatric disorder characterized by appetite and rapid eye movement sleep (REMS) abnormalities. Melanin-concentrating hormone (MCH) and nesfatin-1/NUCB2 (nesfatin) are strongly co - expressed in the hypothalamus and are involved both in food intake regulation and depression. Since MCH was recognized earlier as a hypnogenic factor, we analyzed the potential role of nesfatin on vigilance. DESIGN: We subjected rats to a 72 h-long REMS deprivation using the classic flower pot method, followed by a 3 h-long 'rebound sleep'. Nesfatin mRNA and protein expressions as well as neuronal activity (Fos) were measured by quantitative in situ hybridization technique, ELISA and immunohistochemistry, respectively, in 'deprived' and 'rebound' groups, relative to controls sacrificed at the same time. We also analyzed electroencephalogram of rats treated by intracerebroventricularly administered nesfatin-1, or saline. RESULTS: REMS deprivation downregulated the expression of nesfatin (mRNA and protein), however, enhanced REMS during 'rebound' reversed this to control levels. Additionally, increased transcriptional activity (Fos) was demonstrated in nesfatin neurons during 'rebound'. Centrally administered nesfatin-1 at light on reduced REMS and intermediate stage of sleep, while increased passive wake for several hours and also caused a short-term increase in light slow wave sleep. CONCLUSIONS: The data designate nesfatin as a potential new factor in sleep regulation, which fact can also be relevant in the better understanding of the role of nesfatin in the pathomechanism of depression.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Hypothalamus/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Sleep, REM/drug effects , Wakefulness/drug effects , Animals , Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Electroencephalography , Gene Expression/drug effects , Hypothalamus/physiology , Injections, Intraventricular , Male , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nucleobindins , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep, REM/physiology , Wakefulness/physiology
20.
Psychopharmacology (Berl) ; 228(3): 439-49, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23515582

ABSTRACT

RATIONALE: Selective rapid eye movement sleep (REMS) deprivation using the platform-on-water ("flower pot") method causes sleep rebound with increased REMS, decreased REMS latency, and activation of the melanin-concentrating hormone (MCH) expressing neurons in the hypothalamus. MCH is implicated in the pathomechanism of depression regarding its influence on mood, feeding behavior, and REMS. OBJECTIVES: We investigated the effects of the most selective serotonin reuptake inhibitor escitalopram on sleep rebound following REMS deprivation and, in parallel, on the activation of MCH-containing neurons. METHODS: Escitalopram or vehicle (10 mg/kg, intraperitoneally) was administered to REMS-deprived (72 h) or home cage male Wistar rats. During the 3-h-long "rebound sleep", electroencephalography was recorded, followed by an MCH/Fos double immunohistochemistry. RESULTS: During REMS rebound, the time spent in REMS and the number of MCH/Fos double-labeled neurons in the lateral hypothalamus increased markedly, and REMS latency showed a significant decrease. All these effects of REMS deprivation were significantly attenuated by escitalopram treatment. Besides the REMS-suppressing effects, escitalopram caused an increase in amount of and decrease in latency of slow wave sleep during the rebound. CONCLUSIONS: These results show that despite the high REMS pressure caused by REMS deprivation procedure, escitalopram has the ability to suppress REMS rebound, as well as to diminish the activation of MCH-containing neurons, in parallel. Escitalopram caused a shift from REMS to slow wave sleep during the rebound. Furthermore, these data point to the potential connection between the serotonergic system and MCH in sleep regulation, which can be relevant in depression and in other mood disorders.


Subject(s)
Citalopram/pharmacology , Hypothalamic Hormones/metabolism , Hypothalamus/drug effects , Melanins/metabolism , Neurons/drug effects , Pituitary Hormones/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Sleep, REM/drug effects , Animals , Citalopram/administration & dosage , Electroencephalography , Hypothalamus/metabolism , Hypothalamus/physiopathology , Male , Neurons/metabolism , Rats , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/administration & dosage , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...