1
|
Tóth A, Dobolyi Á. Prolactin in sleep and EEG regulation: New mechanisms and sleep-related brain targets complement classical data. Neurosci Biobehav Rev 2025; 169:106000. [PMID: 39755290 DOI: 10.1016/j.neubiorev.2024.106000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/17/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
The role of prolactin in sleep regulation has been the subject of extensive research over the past 50 years, resulting in the identification of multiple, disparate functions for the hormone. Prolactin demonstrated a characteristic circadian release pattern with elevation during dark and diminution during light. High prolactin levels were linked to non-rapid eye movement sleep and electroencephalogram delta activity in humans. Conversely, hyperprolactinemia showed strong correlation with REM sleep in rodent studies. Prolactin may be implicated in the alterations in female sleep patterns observed during the reproductive cycle, it may play a role in the REM sleep enhancement following stress and in sleep-related immunological processes. In conclusion, prolactin appears to have a sleep-promoting role, particularly during the dark phase. However, it does not appear to play a central and coherent role in sleep regulation, as observed in some neuropeptides such as orexin. Conversely, its principal function may be to facilitate situational, yet adaptive, changes in sleep patterns in response to challenging physiological phases, such as those associated with stress, immunological challenges, or the reproductive cycle. Neuronal substrates for prolactin-mediated sleep effects remain unknown; however, recent rodent sleep studies may provide insights into the potential sites of these effects.
Collapse
Affiliation(s)
- Attila Tóth
- In vivo Electrophysiology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Hungary.
| | - Árpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Hungary
| |
Collapse
|
2
|
Kim M, Kim Y, Lee HW, Kim KM, Kim S, Oh S. The Improvement in Sleep Quality by Zizyphi Semen in Rodent Models Through GABAergic Transmission Regulation. Nutrients 2024; 16:4266. [PMID: 39770888 PMCID: PMC11677272 DOI: 10.3390/nu16244266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Sleep, a process physiologically vital for mental health, faces disruptions in various sleep disorders linked to metabolic and neurodegenerative risks. Zizyphus seed (Zizy) has long been recognized for its diverse pharmacological attributes, including analgesic, sedative, insomnia, and anxiety alleviation. Objectives: In this study, the sleep-prolonging effects of Zizy extract (100, 200 mg/kg), along with their characterizing compounds jujuboside A (JuA) (5, 10 mg/kg), were evaluated in a mouse model under a pentobarbital-induced sleep. Additionally, the efficacy of Zizy extract was examined on caffeine-induced insomnia in mice. Methods: To confirm the efficacy of Zizy extract on the structure and quality of sleep, an electroencephalogram (EEG) analysis of rats was performed using the MATLAB algorithm. Additionally, Western blot analysis and measurement of intracellular chloride influx were performed to confirm whether these effects acted through the gamma-aminobutyric acid (GABA)ergic system. Administration of Zizy extract showed no effect on the locomotor performance of mice, but the extract and their characteristic compounds significantly prolonged sleep duration in comparison to the pentobarbital alone group in the pentobarbital-induced sleep mouse model. Furthermore, this extract alleviated caffeine-induced insomnia in mice. Results: The administration of Zizy extract extended non-rapid eye movement sleep (NREMS) duration without inducing significant changes in the brain wave frequency. Zizy extract regulated the expression of GABAA receptor subunits and GAD65/67 in specific brain regions (frontal cortex, hippocampus, and hypothalamus). JuA increased intracellular chloride influx in human SH-SY5Y cells, and it was reduced by GABAA receptor antagonists. These results suggest that the sleep-maintaining effects of Zizy extract may entail GABAergic regulation. In summary, Zizy extract demonstrated sleep-prolonging properties, improved insomnia, and regulated sleep architecture through GABAergic system modulation. Conclusions: These findings suggest that Zizy extract has potential as a therapeutic agent for stress-related neuropsychiatric conditions such as insomnia.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea;
| | - YuJaung Kim
- Department of Neurology, Ewha Medical Research Institute, Seoul 07804, Republic of Korea; (Y.K.); (H.W.L.)
| | - Hyang Woon Lee
- Department of Neurology, Ewha Medical Research Institute, Seoul 07804, Republic of Korea; (Y.K.); (H.W.L.)
- Department of Neurology, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Kyung-Mi Kim
- Life Science Research Institute, NOVAREX Co., Ltd., Cheongju 28220, Republic of Korea; (K.-M.K.); (S.K.)
| | - Singeun Kim
- Life Science Research Institute, NOVAREX Co., Ltd., Cheongju 28220, Republic of Korea; (K.-M.K.); (S.K.)
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea;
| |
Collapse
|
3
|
Sugumaran R, Sai Krishna KS, Saibaba J, Narayan SK, Sandhiya S, Rajeswari M. Melatonin on sleep in Parkinson's disease: A randomized double blind placebo controlled trial. Sleep Med 2024; 124:502-509. [PMID: 39437460 DOI: 10.1016/j.sleep.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Sleep disturbances are one of the most common non-motor symptoms in Idiopathic Parkinson's Disease (IPD) patients. However, the effect of melatonin on sleep problems in Parkinson's disease patients is unclear. AIMS AND OBJECTIVES To study the effect of melatonin on sleep in IPD patients through subjective and objective assessment. METHODS Between August 2023 to February 2024, we conducted a randomized, double-blind, placebo-controlled trial on IPD patients. We randomized eligible subjects to melatonin (3 mg) (n = 43) or placebo (n = 43) for 8 weeks. The primary endpoint was sleep quality assessed through the Pittsburgh sleep quality index and daytime sleepiness using Epworth sleepiness scale. Secondary endpoints were polysomnographic sleep parameters, quality of life, motor and non-motor symptoms. Assessments were done at baseline and at the end of 8 weeks. RESULTS We screened 107 IPD patients and 86 patients were included in the study. Seventy three patients (melatonin, 35 and placebo, 38) completed the study. The mean change in Pittsburgh Sleep Quality Index (PSQI) score between the two groups was 1.87 (95 % CI: 1.5-2.1; p = 0.001) and Epworth Sleepiness Scale (ESS) score was 1.25 (95 % CI: 0.80-1.71; p = 0.001) favoring melatonin. The mean difference between the two groups for Non-Motor Symptoms Scale (NMSS) was 6.11 (95 % CI 5.27-6.92; p = 0.001), Parkinson's Disease Questionnaire (PDQ 39) 8.12 (95 % CI 6.97-9.50; p = 0.001) & Polysomnography (PSG) parameters [sleep latency 8.36 (95 % CI 4.38-12.34; p = 0.001) and total sleep time 14.51 (95 % CI 5.00-24.41; p = 0.005)] favoring melatonin. Side effects attributable to melatonin were minimal. CONCLUSION Melatonin is an effective and safe treatment option for sleep problems in PD patients, and beneficial effects on sleep quality are associated with improved non-motor symptoms and quality of life. We need to emphasize the fact that though we had statistically significant changes in our outcomes, it is not clear whether such changes would have real-life impact (meaningfulness) that would be relevant to licensing authorities or management as patients in our study are young, have short disease duration, have high use of anticholinergics and on modest levodopa equivalent dose. So, we are doubtful if this could be generalized to the typical PD population who are older, have longer disease duration and are on potentially sedating medications or not.
Collapse
Affiliation(s)
- Ramkumar Sugumaran
- Department of Neurology, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| | - Kadarla Shiva Sai Krishna
- Department of Neurology, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| | - Jayaram Saibaba
- Department of Neurology, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| | - Sunil K Narayan
- Department of Neurology, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| | - S Sandhiya
- Department of Pharmacology, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| | - M Rajeswari
- Department of Biostatistics, Jawaharlal Institute of Post Graduate Medical Education & Research (JIPMER), Puducherry, 605006, India.
| |
Collapse
|
4
|
Fan X, Mao X, Yu P, Han D, Chen C, Wang H, Zhang X, Liu S, Chen W, Chen Z, Du X, Jin L, Song Y, Li H, Zhang N, Wu Y, Chang L, Wang C. Sleep disturbance impaired memory consolidation via lateralized disruption of metabolite in the thalamus and hippocampus: A cross-sectional proton magnetic resonance spectroscopy study. J Alzheimers Dis 2024; 102:1057-1073. [PMID: 39584303 DOI: 10.1177/13872877241295401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
BACKGROUND Memory consolidation in sleep-dependent individuals involves the circuitry connections of cortex, thalamus and hippocampus, regulating via neural metabolites. However, the disruption of metabolic pattern in thalamus and hippocampus remains unclear. OBJECTIVE We aim to explore the disruptive effects of insomnia on the metabolites during memory consolidation, particularly the underlying neurometabolic mechanisms in comorbidity of failed memory consolidation. METHODS This study integrates clinical research with animal experiment. In clinical research, 49 participants were divided into four groups: healthy controls (HC, n = 11), insomnia with normal cognition (IS, n = 14), mild cognitive impairment without insomnia (MCI, n = 10), and insomnia with mild cognitive impairment (IS-MCI, n = 14). Magnetic resonance spectroscopy (MRS) was used to evaluate the neural γ-aminobutyric acid (GABA) and glutamate-glutamine (Glx) in bilateral thalamus. In experimental studies, the rat model of sleep deprivation combined with amyloid-β (Aβ) injection was established, after behavior testing, the levels of Glx, choline (Cho) and N-acetyl aspartate (NAA) in the bilateral hippocampus were evaluated with MRS. RESULTS The patients in the IS-MCI group exhibited significantly lower GABA level than IS, MCI and HC groups. Results from rat studies showed that sleep deprivation exacerbated asymmetric alterations in Aβ-induced bilateral hippocampal metabolite abnormalities, which correlated with cognition. These neuro-metabolite disruption accompanied with synaptic loss and activation of astrocytes. CONCLUSIONS The lateralized decrease in GABA levels of thalamus and NAA, Cho, and Glx levels of hippocampus under conditions of sleep disturbance with cognitive decline may provide evidence for the neural metabolic mechanisms underlying the disruption of memory consolidation.
Collapse
Affiliation(s)
- Xiaowei Fan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Xin Mao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ping Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Ding Han
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Chuxin Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xinyi Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Siyu Liu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Weijing Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
| | - Ziyan Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaoqiang Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Liangyun Jin
- Electron Microscope Room of Central Laboratory, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ning Zhang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Chunxue Wang
- Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Beijing, China
- Department of Neuropsychiatry and Behavioral Neurology and Clinical Psychology, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Ye M, Rheu KM, Lee BJ, Shim I. GABALAGEN Facilitates Pentobarbital-Induced Sleep by Modulating the Serotonergic System in Rats. Curr Issues Mol Biol 2024; 46:11176-11189. [PMID: 39451543 PMCID: PMC11505973 DOI: 10.3390/cimb46100663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/14/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is one of the inhibitory neurotransmitters with beneficial effects including sedative properties. However, despite various clinical trials, scientific evidence regarding the impact on sleep of orally ingested GABA, whether natural or synthesized through biological pathways, is not clear. GABALAGEN (GBL) is the product of fermented collagen by Lactobacillus brevis BJ20 (L. brevis BJ20) and Lactobacillus plantarum BJ21 (L. plantarum BJ21), enriched with GABA and characterized by low molecular weight. The aim of this study was to investigate the effect of GBL on sleep improvement via a receptor binding assay in a pentobarbital-induced sleep-related rat model. We utilized a pentobarbital-induced sleep-related rat model to conduct this research. The present study investigated the sedative effects of GBL through electroencephalography (EEG) analysis in the pentobarbital-induced sleep animal model. Exploration of the neural basis of these positive effects involved evaluating orexin in the brain via immunohistochemical methods and 5-HT in the serum using an enzyme-linked immunosorbent assay (ELISA). Furthermore, we conducted a binding assay for 5-HT2C receptors, as these are considered pivotal targets in the mechanism of action for sleep aids. Diazepam (DZP) was used as a positive control to compare the efficacy of GBL. Results: In the binding assay, GBL displayed binding affinity to the 5-HT2C receptor (IC50 value, 5.911 µg/mL). Administration of a low dose of GBL (GBL_L; 100 mg/kg) increased non-rapid eye movement sleep time and decreased wake time based on EEG data in pentobarbital-induced rats. Administration of a high dose of GBL (GBL_H; 250 mg/kg) increased non-rapid eye movement sleep time. Additionally, GBL groups significantly increased concentration of the 5-HT level in the serum. GBL_H decreased orexin expression in the lateral hypothalamus. Conclusion: Overall, the sedative effect of GBL may be linked to the activation of serotonergic systems, as indicated by the heightened affinity of the 5-HT2C receptor binding and elevated levels of 5-HT observed in the serum. This suggests that GBL holds promise as a novel compound for inducing sleep in natural products.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Kyoung-min Rheu
- Marine Bioprocess Co., Ltd., Busan 46048, Republic of Korea; (K.-m.R.); (B.-j.L.)
| | - Bae-jin Lee
- Marine Bioprocess Co., Ltd., Busan 46048, Republic of Korea; (K.-m.R.); (B.-j.L.)
| | - Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| |
Collapse
|
6
|
Cheaha D, Basor N, Manor R, Hayeemasae N, Samerphob N. Sedative and hypnotic effects with cortical EEG sleep-wake profiles of Millingtonia hortensis dried flower aqueous in mice. Heliyon 2024; 10:e37531. [PMID: 39296192 PMCID: PMC11409071 DOI: 10.1016/j.heliyon.2024.e37531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024] Open
Abstract
The ethnopharmacological relevance of the Millingtonia hortensis (M. hortensis) flower's aqueous extract lies in its traditional use as a herbal remedy in Southeast Asia. With a rich history in folk medicine, this aqueous has been esteemed for its purported sedative and anxiolytic properties. Our research delves into the scientific basis of these traditional claims, exploring the potential mechanisms underlying the observed effects of M. hortensis flower's aqueous extract on sleep promotion and mood regulation. This study aimed to explore the sleep-promoting effects of M. hortensis dried flower in mice, using an aqueous concentration equivalent to a human concentration of 2.7 mg/mL. Anxiolytic and antidepressant properties were evaluated using behavioural tests, while electroencephalography (EEG) analysis probed the neural mechanisms underlying sleep promotion post-consumption. The aqueous extract of M. hortensis dried flower administered to mice showed a decrease in immobility in the forced swimming test, demonstrating antidepressant-like effects. Moreover, mice treated with M. hortensis aqueous exhibited increased non-rapid eye movement (NREM) sleep duration, corroborating sleep-promoting potential. EEG analysis of mice treated with M. hortensis aqueous revealed heightened beta oscillations in the frontal and parietal cortex, while pre-treatment with M. hortensis aqueous or diazepam enhanced rapid eye movement (REM) sleep after thiopental administration. Interestingly, M. hortensis aqueous pre-treatment augmented delta frequency ranges in the frontal cortex. Overall, these findings indicate that M. hortensis dried flower's aqueous extract, at a human-equivalent dosage, exerts significant behavioural and neural effects specifically, sedative and hypnotic aspects in mice, corroborating its potential as a natural remedy to promote sleep and regulate mood.
Collapse
Affiliation(s)
- Dania Cheaha
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Thailand
| | - Nurulhuda Basor
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Thailand
| | - Rodiya Manor
- Faculty of Science and Technology, Prince of Songkla University, Thailand
| | - Nabil Hayeemasae
- Faculty of Science and Technology, Prince of Songkla University, Thailand
| | - Nifareeda Samerphob
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Thailand
| |
Collapse
|
7
|
Lee DH, Jee HJ, Jung YS. β-Lapachone Exerts Hypnotic Effects via Adenosine A 1 Receptor in Mice. Biomol Ther (Seoul) 2024; 32:531-539. [PMID: 39164983 PMCID: PMC11392670 DOI: 10.4062/biomolther.2024.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/03/2024] [Accepted: 08/03/2024] [Indexed: 08/22/2024] Open
Abstract
Sleep is one of the most essential physiological phenomena for maintaining health. Sleep disturbances, such as insomnia, are often accompanied by psychiatric or physical conditions such as impaired attention, anxiety, and stress. Medication used to treat insomnia have concerns about potential side effects with long-term use, so interest in the use of alternative medicine is increasing. In this study, we investigated the hypnotic effects of β-lapachone (β-Lap), a natural naphthoquinone compound, using pentobarbital-induced sleep test, immunohistochemistry, real-time PCR, and western blot in mice. Our results indicated that β-Lap exerts a significant hypnotic effect by showing a decrease in sleep onset latency and an increase in total sleep time in pentobarbital-induced sleep model. The results of c-Fos immunostaining showed that β-Lap decreased neuronal activity in the basal forebrain and lateral hypothalamus, which are wakefulness-promoting brain regions, while increasing in the ventrolateral preoptic nucleus, a sleep-promoting region; all these effects were significantly abolished by 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), an adenosine A1 receptor (A1R) antagonist. Western blot analysis showed that β-Lap increased extracellular signalregulated kinase phosphorylation and nuclear factor-kappa B translocation from the cytoplasm to the nucleus; these effects were inhibited by DPCPX. Additionally, β-Lap increased the mRNA levels of A1R. Taken together, these results suggest that β-Lap exerts hypnotic effects, potentially through A1R.
Collapse
Affiliation(s)
- Do Hyun Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Hye Jin Jee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
8
|
Kim YS, Lee BK, Kim CS, Lee YS, Lee YJ, Kim KW, Lee DY, Jung YS. Sedum kamtschaticum Exerts Hypnotic Effects via the Adenosine A 2A Receptor in Mice. Nutrients 2024; 16:2611. [PMID: 39203748 PMCID: PMC11357430 DOI: 10.3390/nu16162611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Insomnia is a common sleep disorder with significant societal and economic impacts. Current pharmacotherapies for insomnia are often accompanied by side effects, necessitating the development of new therapeutic drugs. In this study, the hypnotic effects and mechanisms of Sedum kamtschaticum 30% ethanol extract (ESK) and one of its active compounds, myricitrin, were investigated using pentobarbital-induced sleep experiments, immunohistochemistry (IHC), receptor binding assays, and enzyme-linked immunosorbent assay (ELISA). The pentobarbital-induced sleep experiments revealed that ESK and myricitrin reduced sleep latency and prolonged total sleep time in a dose-dependent manner. Based on c-Fos immunostaining, ESK, and myricitrin enhanced the GABAergic neural activity in sleep-promoting ventrolateral preoptic nucleus (VLPO) GABAergic. By measuring the level of GABA released from VLPO GABAergic neurons, ESK and myricitrin were found to increase GABA release in the hypothalamus. These effects were significantly inhibited by SCH. Moreover, ESK exhibited a concentration-dependent binding affinity for the adenosine A2A receptors (A2AR). In conclusion, ESK and myricitrin have hypnotic effects, and their underlying mechanisms may be related to the activation of A2AR.
Collapse
Affiliation(s)
- Yeon-Soo Kim
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (Y.-S.K.); (B.K.L.)
| | - Bo Kyung Lee
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (Y.-S.K.); (B.K.L.)
| | - Cha Soon Kim
- Research and Development Department, Genencell Co., Ltd., Yongin 16950, Republic of Korea;
| | - Young-Seob Lee
- Development of Herbal Crop Research, Rural Development Administration (RDA), Eumseong 27709, Republic of Korea; (Y.-S.L.); (Y.J.L.); (K.-W.K.)
| | - Yoon Ji Lee
- Development of Herbal Crop Research, Rural Development Administration (RDA), Eumseong 27709, Republic of Korea; (Y.-S.L.); (Y.J.L.); (K.-W.K.)
| | - Kwan-Woo Kim
- Development of Herbal Crop Research, Rural Development Administration (RDA), Eumseong 27709, Republic of Korea; (Y.-S.L.); (Y.J.L.); (K.-W.K.)
| | - Dae Young Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Yi-Sook Jung
- Department of Pharmacy, Ajou University, Suwon 16499, Republic of Korea; (Y.-S.K.); (B.K.L.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
9
|
Zheng J, Wu M, Pang Y, Liu Q, Liu Y, Jin X, Tang J, Bao L, Niu Y, Zheng Y, Zhang R. Interior decorative volatile organic compounds exposure induces sleep disorders through aberrant branched chain amino acid transaminase 2 mediated glutamatergic signaling resulting from a neuroinflammatory cascade. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 934:173254. [PMID: 38761924 DOI: 10.1016/j.scitotenv.2024.173254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/16/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Air pollution has been recognized as a contributing factor to sleep disorders (SD), which have been correlated with an elevated susceptibility to a variety of human diseases. Nevertheless, research has not definitively established a connection between SD and interior decorative volatile organic compounds (ID-VOCs), a significant indoor air pollutant. In this study, we employed a mouse model exposed to ID-VOCs to explore the impacts of ID-VOCs exposure on sleep patterns and the potential underlying mechanism. Of the 23 key compositions of ID-VOCs identified, aromatic hydrocarbons were found to be the most prevalent. Exposure to ID-VOCs in mice resulted in SD, characterized by prolonged wake fullness and decreased sleep during the light period. ID-VOCs exposure triggered neuroinflammatory responses in the suprachiasmatic nucleus (SCN), with microglia activation leading to the overproduction of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1α (IL-1α), and complement component 1q (C1q), ultimately inducing A1 astrocytes. Consequently, the upregulation of branched chain amino acid transaminase 2 (BCAT2) in A1 astrocytes resulted in elevated extracellular glutamate and disruption of the wake-sleep transition mechanism, which might be the toxicological mechanism of SD caused by ID-VOCs.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, PR China
| | - Mengqi Wu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yaxian Pang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Qingping Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yan Liu
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; School of Public Health, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia, PR China
| | - Xiaoting Jin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, Shandong, PR China
| | - Jinglong Tang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, Shandong, PR China
| | - Lei Bao
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yujie Niu
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, Shandong, PR China.
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China.
| |
Collapse
|
10
|
Wu Y, Tang Z, Du S, Dai J, Shen J, Niu H, Song X, Huang G, Du L, Jin Y. Oral quercetin nanoparticles in hydrogel microspheres alleviate high-altitude sleep disturbance based on the gut-brain axis. Int J Pharm 2024; 658:124225. [PMID: 38750982 DOI: 10.1016/j.ijpharm.2024.124225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
High-altitude sleep disturbance is a common symptom of acute mountain sickness, which can be alleviated via modulation of the gut-brain axis. Quercetin (Que) is used to modulate gut microbiota and serves as a potential drug to regulate the gut-brain axis, but the poor solubility and bioavailability affect its biological functions. Here, Que nanoparticles (QNPs) were prepared with zein using an antisolvent method, and QNP-loaded calcium alginate hydrogel microspheres (QNP@HMs) were prepared using electrospinning technology to improve the gastrointestinal stability and intestinal adhesion of QNPs. In the mouse model of high-altitude sleep disturbance, oral administration of QNP@HMs before the mice entering high altitude prolonged sleep duration, improved blood cell recovery, spontaneous behavior and short-term memory, and reduced such inflammation factors as TNF-α and iNOS. Moreover, QNP@HMs enhanced the abundance of probiotics in the gut, including Lactobacillus and Lachnospira, and reduced intestinal inflammation. However, in the mice after gut sterilization by long-term oral antibiotics, QNP@HMs showed no therapeutic effect. QNP@HMs are a promising medication for the prevention of high-altitude sleep disturbance based on the gut-brain axis.
Collapse
Affiliation(s)
- Yanping Wu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ziyan Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Shumin Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing Dai
- Department of Information, General Hospital of Western Theater Command of PLA, Chengdu 610083, China
| | - Jintao Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hong Niu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xingshuang Song
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Guiyu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lina Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
11
|
Shiromani PJ, Vidal-Ortiz A. Most dynorphin neurons in the zona incerta-perifornical area are active in waking relative to non-rapid-eye movement and rapid-eye movement sleep. Sleep 2024; 47:zsae065. [PMID: 38447008 PMCID: PMC11494376 DOI: 10.1093/sleep/zsae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/26/2024] [Indexed: 03/08/2024] Open
Abstract
Dynorphin is an endogenous opiate localized in many brain regions and spinal cord, but the activity of dynorphin neurons during sleep is unknown. Dynorphin is an inhibitory neuropeptide that is coreleased with orexin, an excitatory neuropeptide. We used microendoscopy to test the hypothesis that, like orexin, the dynorphin neurons are wake-active. Dynorphin-cre mice (n = 3) were administered rAAV8-Ef1a-Con/Foff 2.0-GCaMP6M into the zona incerta-perifornical area, implanted with a GRIN lens (gradient reflective index), and electrodes to the skull that recorded sleep. One month later, a miniscope imaged calcium fluorescence in dynorphin neurons during multiple bouts of wake, non-rapid-eye movement (NREM), and rapid-eye movement (REM) sleep. Unbiased data analysis identified changes in calcium fluorescence in 64 dynorphin neurons. Most of the dynorphin neurons (72%) had the highest fluorescence during bouts of active and quiet waking compared to NREM or REM sleep; a subset (20%) were REM-max. Our results are consistent with the emerging evidence that the activity of orexin neurons can be classified as wake-max or REM-max. Since the two neuropeptides are coexpressed and coreleased, we suggest that dynorphin-cre-driven calcium sensors could increase understanding of the role of this endogenous opiate in pain and sleep.
Collapse
Affiliation(s)
- Priyattam J Shiromani
- Laboratory of Sleep Medicine and Chronobiology, Research Service, Ralph H. Johnson Veterans Healthcare System Charleston, SC, USA
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Aurelio Vidal-Ortiz
- Laboratory of Sleep Medicine and Chronobiology, Research Service, Ralph H. Johnson Veterans Healthcare System Charleston, SC, USA
| |
Collapse
|
12
|
Wang Y, Wang Y, Liu Y, Cheng H, Dagnew TM, Xu Y, Wang C. Synthesis and Characterization of a New Carbon-11 Labeled Positron Emission Tomography Radiotracer for Orexin 2 Receptors Neuroimaging. Drug Des Devel Ther 2024; 18:215-222. [PMID: 38312991 PMCID: PMC10838518 DOI: 10.2147/dddt.s404992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/13/2023] [Indexed: 02/06/2024] Open
Abstract
Purpose Orexin receptors (OXRs) play a crucial role in modulating various physiological and neuropsychiatric functions within the central nervous system (CNS). Despite their significance, the precise role of OXRs in the brain remains elusive. Positron emission tomography (PET) imaging is instrumental in unraveling CNS functions, and the development of specific PET tracers for OXRs is a current research focus. Methods The study investigated MDK-5220, an OX2R-selective agonist with promising binding properties (EC50 on OX2R: 0.023 μM, Ki on hOX2R: 0.14 μM). Synthesized and characterized as an OX2R PET probe, [11C]MDK-5220 was evaluated for its potential as a tracer. Biodistribution studies in mice were conducted to assess OX2R binding selectivity, with particular attention to its interaction with P-glycoprotein (P-gp) on the blood-brain barrier. Results [11C]MDK-5220 exhibited promising attributes as an OX2R PET probe, demonstrating robust OX2R binding selectivity in biodistribution studies. However, an observed interaction with P-gp impacted its brain uptake. Despite this limitation, [11C]MDK-5220 presents itself as a potential candidate for further development. Discussion The study provides insights into the functionality of the OX system and the potential of [11C]MDK-5220 as an OX2R PET probe. The observed interaction with P-gp highlights a consideration for future modifications to enhance brain uptake. The findings pave the way for innovative tracer development and propel ongoing research on OX systems, contributing to a deeper understanding of their role in the CNS. Conclusion [11C]MDK-5220 emerges as a promising OX2R PET probe, despite challenges related to P-gp interaction. This study lays the foundation for further exploration and development of PET probes targeting OXRs, opening avenues for advancing our understanding of OX system functionality within the brain.
Collapse
Affiliation(s)
- Yanli Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yongle Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- School of Pharmacy, Minzu University of China, Beijing, 100081, People’s Republic of China
| | - Yan Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Hua Cheng
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Tewodros Mulugeta Dagnew
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yulong Xu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| |
Collapse
|
13
|
Maldonado PE, Concha-Miranda M, Schwalm M. Autogenous cerebral processes: an invitation to look at the brain from inside out. Front Neural Circuits 2023; 17:1253609. [PMID: 37941893 PMCID: PMC10629273 DOI: 10.3389/fncir.2023.1253609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
While external stimulation can reliably trigger neuronal activity, cerebral processes can operate independently from the environment. In this study, we conceptualize autogenous cerebral processes (ACPs) as intrinsic operations of the brain that exist on multiple scales and can influence or shape stimulus responses, behavior, homeostasis, and the physiological state of an organism. We further propose that the field should consider exploring to what extent perception, arousal, behavior, or movement, as well as other cognitive functions previously investigated mainly regarding their stimulus-response dynamics, are ACP-driven.
Collapse
Affiliation(s)
- Pedro E. Maldonado
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile
- National Center for Artificial Intelligence (CENIA), Santiago, Chile
| | - Miguel Concha-Miranda
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Miriam Schwalm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
14
|
Showler L, Ali Abdelhamid Y, Goldin J, Deane AM. Sleep during and following critical illness: A narrative review. World J Crit Care Med 2023; 12:92-115. [PMID: 37397589 PMCID: PMC10308338 DOI: 10.5492/wjccm.v12.i3.92] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 03/22/2023] [Indexed: 06/08/2023] Open
Abstract
Sleep is a complex process influenced by biological and environmental factors. Disturbances of sleep quantity and quality occur frequently in the critically ill and remain prevalent in survivors for at least 12 mo. Sleep disturbances are associated with adverse outcomes across multiple organ systems but are most strongly linked to delirium and cognitive impairment. This review will outline the predisposing and precipitating factors for sleep disturbance, categorised into patient, environmental and treatment-related factors. The objective and subjective methodologies used to quantify sleep during critical illness will be reviewed. While polysomnography remains the gold-standard, its use in the critical care setting still presents many barriers. Other methodologies are needed to better understand the pathophysiology, epidemiology and treatment of sleep disturbance in this population. Subjective outcome measures, including the Richards-Campbell Sleep Questionnaire, are still required for trials involving a greater number of patients and provide valuable insight into patients’ experiences of disturbed sleep. Finally, sleep optimisation strategies are reviewed, including intervention bundles, ambient noise and light reduction, quiet time, and the use of ear plugs and eye masks. While drugs to improve sleep are frequently prescribed to patients in the ICU, evidence supporting their effectiveness is lacking.
Collapse
Affiliation(s)
- Laurie Showler
- Intensive Care Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| | - Yasmine Ali Abdelhamid
- Intensive Care Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| | - Jeremy Goldin
- Sleep and Respiratory Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| | - Adam M Deane
- Intensive Care Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| |
Collapse
|
15
|
Ruiz-Viroga V, de Ceglia M, Morelli L, Castaño EM, Calvo EB, Suárez J, Rodríguez de Fonseca F, Galeano P, Lagos P. Acute intrahippocampal administration of melanin-concentrating hormone impairs memory consolidation and decreases the expression of MCHR-1 and TrkB receptors. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110703. [PMID: 36565982 DOI: 10.1016/j.pnpbp.2022.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Interest in the role of melanin-concentrating hormone (MCH) in memory processes has increased in recent years, with some studies reporting memory-enhancing effects, while others report deleterious effects. Due to these discrepancies, this study seeks to provide new evidence about the role of MCH in memory consolidation and its relation with BDNF/TrkB system. To this end, in the first experiment, increased doses of MCH were acutely administered in both hippocampi to groups of male rats (25, 50, 200, and 500 ng). Microinjections were carried out immediately after finishing the sample trial of two hippocampal-dependent behavioral tasks: the Novel Object Recognition Test (NORT) and the modified Elevated Plus Maze (mEPM) test. Results indicated that a dose of 200 ng of MCH or higher impaired memory consolidation in both tasks. A second experiment was performed in which a dose of 200 ng of MCH was administered alone or co-administered with the MCHR-1 antagonist ATC-0175 at the end of the sample trial in the NORT. Results showed that MCH impaired memory consolidation, while the co-administration with ATC-0175 reverted this detrimental effect. Moreover, MCH induced a significant decrease in hippocampal MCHR-1 and TrkB expression with no modification in the expression of BDNF and NMDA receptor subunits NR1, NR2A, and NR2B. These results suggest that MCH in vivo elicits pro-amnesic effects in the rat hippocampus by decreasing the availability of its receptor and TrkB receptors, thus linking both endogenous systems to memory processes.
Collapse
Affiliation(s)
- Vicente Ruiz-Viroga
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay
| | - Marialuisa de Ceglia
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo M Castaño
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Eduardo Blanco Calvo
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Campus de Teatinos S/N, Málaga 29071, Spain.
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, Málaga 29071, Spain.
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, Av. Carlos Haya 82, Málaga 29010, Spain.
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina.
| | - Patricia Lagos
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, Montevideo ZP11800, Uruguay.
| |
Collapse
|
16
|
Kim M, Kim Y, Lee HW, Jung JC, Oh S. Chrysanthemum morifolium and Its Bioactive Substance Enhanced the Sleep Quality in Rodent Models via Cl - Channel Activation. Nutrients 2023; 15:1309. [PMID: 36986039 PMCID: PMC10059900 DOI: 10.3390/nu15061309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Dried Chrysanthemum morifolium (Chry) flowers have been used in Korea as a traditional insomnia treatment. In this study, the sleep-promoting activity and improving sleep quality of Chry extract (ext) and its active substance linarin were analyzed by pentobarbital-induced sleep experiment in mice and electroencephalography (EEG), electromyogram (EMG) analysis in rats. In a dose-dependent manner, Chry ext and linarin promoted longer sleep duration in the pentobarbital-induced sleep test compared to pentobarbital-only groups at both hypnotic and subhypnotic doses. Chry ext administration also significantly improved sleep quality, as seen in the relative power of low-frequency (delta) waves when compared with the control group. Linarin increased Cl- uptake in the SH-SY5Y human cell line and chloride influx was reduced by bicuculline. After administration of Chry ext, the hippocampus, frontal cortex, and hypothalamus from rodents were collected and blotted for glutamic acid decarboxylase (GAD)65/67 and gamma-aminobutyric acid (GABA)A receptors subunit expression levels. The expression of α1-subunits, β2-subunits, and GAD65/67 of the GABAA receptor was modulated in the rodent brain. In conclusion, Chry ext augments pentobarbital-induced sleep duration and enhances sleep quality in EEG waves. These effects might be due to the activation of the Cl- channel.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - YuJaung Kim
- Department of Neurology, Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hyang Woon Lee
- Department of Neurology, Medical Research Institute, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
- Graduate Programs in Artificial Intelligence Convergence, Computational Medicine, System Health Science and Engineering, Ewha Womans University, Seoul 03765, Republic of Korea
| | - Jae-Chul Jung
- Life Science Research Institute, NOVAREX Co., Ltd., Cheongju 28220, Republic of Korea
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|
17
|
BaHammam AS, Pirzada AR, Pandi-Perumal SR. Neurocognitive, mood changes, and sleepiness in patients with REM-predominant obstructive sleep apnea. Sleep Breath 2023; 27:57-66. [PMID: 35318576 DOI: 10.1007/s11325-022-02602-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/13/2022] [Accepted: 03/17/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE This article focuses on recent evidence linking rapid eye movement (REM) obstructive sleep apnea (OSA) (REM-OSA) to neurocognitive dysfunction and mood changes; the proposed mechanisms for increased risk of neurocognitive dysfunction in REM-OSA, and future research prospects. METHODS PubMed and Google Scholar records were examined for articles utilizing pre-defined keywords. In this work, we mainly included studies published after 2017; nevertheless, critical studies published prior to 2017 were considered. RESULTS REM-OSA is an under-recognized stage-related sleep-disordered breathing in which obstructive respiratory events happen chiefly in stage REM. The disorder is commonly seen amongst younger patients and females and has recently been linked to cardiometabolic complications. Although less symptomatic than non-REM-OSA and non-stage-specific OSA, current findings indicate that REM-OSA may have neurocognitive repercussions and mood changes and could be linked to insomnia, increased dreams, and nightmares. CONCLUSION Currently available evidence indicates that REM-OSA may present with insomnia and nightmares and could affect cognitive function and mood.
Collapse
Affiliation(s)
- Ahmed S BaHammam
- Department of Medicine, The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia. .,Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi, Arabia (08-MED511-02), Riyadh, Saudi Arabia.
| | - Abdul Rouf Pirzada
- Department of Medicine, The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,North Cumbria Integrated Care (NCIC), NHS, Carlisle, UK
| | - Seithikurippu R Pandi-Perumal
- Department of Medicine, The University Sleep Disorders Center, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
18
|
Albinni B, de Zambotti M, Iacovides S, Baker FC, King CD. The complexities of the sleep-pain relationship in adolescents: A critical review. Sleep Med Rev 2023; 67:101715. [PMID: 36463709 PMCID: PMC9868111 DOI: 10.1016/j.smrv.2022.101715] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/20/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022]
Abstract
Chronic pain is a common and disabling condition in adolescents. Disturbed sleep is associated with many detrimental effects in adolescents with acute and chronic pain. While sleep and pain are known to share a reciprocal relationship, the sleep-pain relationship in adolescence warrants further contextualization within normally occurring maturation of several biopsychological processes. Since sleep and pain disorders begin to emerge in early adolescence and are often comorbid, there is a need for a comprehensive picture of their interrelation especially related to temporal relationships and mechanistic drivers. While existing reviews provide a solid foundation for the interaction between disturbed sleep and pain in youth, we will extend this review by highlighting current methodological challenges for both sleep and pain assessments, exploring the recent evidence for directionality in the sleep-pain relationship, reviewing potential mechanisms and factors underlying the relationship, and providing direction for future investigations. We will also highlight the potential role of digital technologies in advancing the understanding of the sleep and pain relationship. Ultimately, we anticipate this information will facilitate further research and inform the management of pain and poor sleep, which will ultimately improve the quality of life in adolescents and reduce the risk of pain persisting into adulthood.
Collapse
Affiliation(s)
- Benedetta Albinni
- Center for Health Sciences, SRI International, Menlo Park, CA, USA; Department of Psychology, University of Campania "Luigi Vanvitelli", Italy
| | | | - Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Center for Health Sciences, SRI International, Menlo Park, CA, USA; Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Christopher D King
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Behavioral Medicine and Clinical Psychology, Pediatric Pain Research Center (PPRC), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Creatura GM, Ered A, Murty VP, Ellman LM. The relationship between sleep, dissociation and psychotic-like experiences. Early Interv Psychiatry 2022; 16:1353-1358. [PMID: 35333007 DOI: 10.1111/eip.13284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/02/2022] [Accepted: 03/13/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Sleep disturbances have frequently been associated with the full spectrum of psychosis, from psychotic-like experiences (PLEs) to individuals who meet diagnostic criteria for schizophrenia. Similarly, dissociative experiences have been linked to both sleep disturbances and PLEs. AIM The aim of this study was to examine the role of dissociation in the relationship between sleep quality and PLEs. METHODS PLEs, dissociative symptoms, and sleep quality were examined in 1677 young adults using self-report measures. A mediation analysis was performed to examine whether dissociative experiences account for some of the relationship between sleep quality and PLEs. RESULTS Dissociative symptoms significantly mediated the relationship between sleep quality and PLEs, with both age and gender used as covariates. CONCLUSION These findings suggest that dissociation may be a key contributor to the relationship between disrupted sleep and PLEs, which could have treatment and identification implications.
Collapse
Affiliation(s)
- Gina M Creatura
- Department of Psychology, Temple University, Philadelphia, Pennsylvania, USA
| | - Arielle Ered
- Department of Psychology, Temple University, Philadelphia, Pennsylvania, USA
| | - Vishnu P Murty
- Department of Psychology, Temple University, Philadelphia, Pennsylvania, USA
| | - Lauren M Ellman
- Department of Psychology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Wang T, Wang X, Tian Y, Gang W, Li X, Yan J, Yuan Y. Modulation effect of low-intensity transcranial ultrasound stimulation on REM and NREM sleep. Cereb Cortex 2022; 33:5238-5250. [PMID: 36376911 DOI: 10.1093/cercor/bhac413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Previous studies have shown that modulating neural activity can affect rapid eye movement (REM) and non-rapid eye movement (NREM) sleep. Low-intensity transcranial ultrasound stimulation (TUS) can effectively modulate neural activity. However, the modulation effect of TUS on REM and NREM sleep is still unclear. In this study, we used ultrasound to stimulate motor cortex and hippocampus, respectively, and found the following: (i) In healthy mice, TUS increased the NREM sleep ratio and decreased the REM sleep ratio, and altered the relative power and sample entropy of the delta band and spindle in NREM sleep and that of the theta and gamma bands in REM sleep. (ii) In sleep-deprived mice, TUS decreased the ratio of REM sleep or the relative power of the theta band during REM sleep. (iii) In sleep-disordered Alzheimer’s disease (AD) mice, TUS increased the total sleep time and the ratio of NREM sleep and modulated the relative power and the sample entropy of the delta and spindle bands during NREM and that of the theta band during REM sleep. These results demonstrated that TUS can effectively modulate REM and NREM sleep and that modulation effect depends on the sleep state of the samples, and can improve sleep in sleep-disordered AD mice.
Collapse
Affiliation(s)
- Teng Wang
- Yanshan University School of Electrical Engineering, , Qinhuangdao 066004 , China
- Yanshan University Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, , Qinhuangdao 066004 , China
| | - Xingran Wang
- Yanshan University School of Electrical Engineering, , Qinhuangdao 066004 , China
- Yanshan University Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, , Qinhuangdao 066004 , China
| | - Yanfei Tian
- Hebei Medical University Department of Pharmacology, , Shijiazhuang, Hebei 050017 , China
| | - Wei Gang
- Hebei Medical University Department of Pharmacology, , Shijiazhuang, Hebei 050017 , China
| | - Xiaoli Li
- Beijing Normal University State Key Laboratory of Cognitive Neuroscience and Learning, , Beijing 100875 , China
| | - Jiaqing Yan
- North China University of Technology College of Electrical and Control Engineering, , Beijing 100041 , China
| | - Yi Yuan
- Yanshan University School of Electrical Engineering, , Qinhuangdao 066004 , China
- Yanshan University Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, , Qinhuangdao 066004 , China
| |
Collapse
|
21
|
Dissel S, Klose MK, van Swinderen B, Cao L, Ford M, Periandri EM, Jones JD, Li Z, Shaw PJ. Sleep-promoting neurons remodel their response properties to calibrate sleep drive with environmental demands. PLoS Biol 2022; 20:e3001797. [PMID: 36173939 PMCID: PMC9521806 DOI: 10.1371/journal.pbio.3001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/16/2022] [Indexed: 01/29/2023] Open
Abstract
Falling asleep at the wrong time can place an individual at risk of immediate physical harm. However, not sleeping degrades cognition and adaptive behavior. To understand how animals match sleep need with environmental demands, we used live-brain imaging to examine the physiological response properties of the dorsal fan-shaped body (dFB) following interventions that modify sleep (sleep deprivation, starvation, time-restricted feeding, memory consolidation) in Drosophila. We report that dFB neurons change their physiological response-properties to dopamine (DA) and allatostatin-A (AstA) in response to different types of waking. That is, dFB neurons are not simply passive components of a hard-wired circuit. Rather, the dFB neurons intrinsically regulate their response to the activity from upstream circuits. Finally, we show that the dFB appears to contain a memory trace of prior exposure to metabolic challenges induced by starvation or time-restricted feeding. Together, these data highlight that the sleep homeostat is plastic and suggests an underlying mechanism.
Collapse
Affiliation(s)
- Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (SD); (PJS)
| | - Markus K. Klose
- University of Pittsburgh School of Medicine, Department of Pharmacology & Chemical Biology, Pittsburgh, Pennsylvania, United States of America
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
| | - Lijuan Cao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Melanie Ford
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Erica M. Periandri
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph D. Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Zhaoyi Li
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul J. Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (SD); (PJS)
| |
Collapse
|
22
|
Oh JY, Walsh CM, Ranasinghe K, Mladinov M, Pereira FL, Petersen C, Falgàs N, Yack L, Lamore T, Nasar R, Lew C, Li S, Metzler T, Coppola Q, Pandher N, Le M, Heuer HW, Heinsen H, Spina S, Seeley WW, Kramer J, Rabinovici GD, Boxer AL, Miller BL, Vossel K, Neylan TC, Grinberg LT. Subcortical Neuronal Correlates of Sleep in Neurodegenerative Diseases. JAMA Neurol 2022; 79:498-508. [PMID: 35377391 PMCID: PMC8981071 DOI: 10.1001/jamaneurol.2022.0429] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022]
Abstract
Importance Sleep disturbance is common among patients with neurodegenerative diseases. Examining the subcortical neuronal correlates of sleep disturbances is important to understanding the early-stage sleep neurodegenerative phenomena. Objectives To examine the correlation between the number of important subcortical wake-promoting neurons and clinical sleep phenotypes in patients with Alzheimer disease (AD) or progressive supranuclear palsy (PSP). Design, Setting, and Participants This longitudinal cohort study enrolled 33 patients with AD, 20 patients with PSP, and 32 healthy individuals from the Memory and Aging Center of the University of California, San Francisco, between August 22, 2008, and December 31, 2020. Participants received electroencephalographic and polysomnographic sleep assessments. Postmortem neuronal analyses of brainstem hypothalamic wake-promoting neurons were performed and were included in the clinicopathological correlation analysis. No eligible participants were excluded from the study. Exposures Electroencephalographic and polysomnographic assessment of sleep and postmortem immunohistological stereological analysis of 3 wake-promoting nuclei (noradrenergic locus coeruleus [LC], orexinergic lateral hypothalamic area [LHA], and histaminergic tuberomammillary nucleus [TMN]). Main Outcomes and Measures Nocturnal sleep variables, including total sleep time, sleep maintenance, rapid eye movement (REM) latency, and time spent in REM sleep and stages 1, 2, and 3 of non-REM (NREM1, NREM2, and NREM3, respectively) sleep, and wake after sleep onset. Neurotransmitter, tau, and total neuronal counts of LC, LHA, and TMN. Results Among 19 patients included in the clinicopathological correlation analysis, the mean (SD) age at death was 70.53 (7.75) years; 10 patients (52.6%) were female; and all patients were White. After adjusting for primary diagnosis, age, sex, and time between sleep analyses and death, greater numbers of LHA and TMN neurons were correlated with decreased homeostatic sleep drive, as observed by less total sleep time (LHA: r = -0.63; P = .009; TMN: r = -0.62; P = .008), lower sleep maintenance (LHA: r = -0.85; P < .001; TMN: r = -0.78; P < .001), and greater percentage of wake after sleep onset (LHA: r = 0.85; P < .001; TMN: r = 0.78; P < .001). In addition, greater numbers of LHA and TMN neurons were correlated with less NREM2 sleep (LHA: r = -0.76; P < .001; TMN: r = -0.73; P < .001). A greater number of TMN neurons was also correlated with less REM sleep (r = -0.61; P = .01). A greater number of LC neurons was mainly correlated with less total sleep time (r = -0.68; P = .008) and greater REM latency (r = 0.71; P = .006). The AD-predominant group had significantly greater sleep drive, including higher total sleep time (mean [SD], 0.49 [1.18] vs -1.09 [1.37]; P = .03), higher sleep maintenance (mean [SD], 0.18 [1.22] vs -1.53 [1.78]; P = .02), and lower percentage of wake after sleep onset during sleep period time (mean [SD], -0.18 [1.20] vs 1.49 [1.72]; P = .02) than the PSP-predominant group based on unbiased k-means clustering and principal component analyses. Conclusions and Relevance In this cohort study, subcortical wake-promoting neurons were significantly correlated with sleep phenotypes in patients with AD and PSP, suggesting that the loss of wake-promoting neurons among patients with neurodegenerative conditions may disturb the control of sleep-wake homeostasis. These findings suggest that the subcortical system is a primary mechanism associated with sleep disturbances in the early stages of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jun Y. Oh
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- School of Medicine, University of California, San Francisco, San Francisco
| | - Christine M. Walsh
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Kamalini Ranasinghe
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Mihovil Mladinov
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Felipe L. Pereira
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Cathrine Petersen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Neus Falgàs
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
| | - Leslie Yack
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
| | - Tia Lamore
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Rakin Nasar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Caroline Lew
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Song Li
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Thomas Metzler
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
| | - Quentin Coppola
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Natalie Pandher
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Michael Le
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Hilary W. Heuer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Helmut Heinsen
- Department of Psychiatry, University of Wurzburg, Wurzburg, Germany
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - William W. Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Joel Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Gil D. Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
| | - Keith Vossel
- Department of Neurology, University of California, Los Angeles, Los Angeles
| | - Thomas C. Neylan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Stress and Health Research Program, Department of Mental Health, San Francisco VA Medical Center, San Francisco, California
- Department of Psychiatry, University of California, San Francisco, San Francisco
| | - Lea T. Grinberg
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco
- Global Brain Health Institute, University of California, San Francisco, San Francisco
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
- Department of Pathology, University of California, San Francisco, San Francisco
| |
Collapse
|
23
|
Wei Y, Xu J, Miao S, Wei K, Peng L, Wang Y, Wei X. Recent advances in the utilization of tea active ingredients to regulate sleep through neuroendocrine pathway, immune system and intestinal microbiota. Crit Rev Food Sci Nutr 2022; 63:7598-7626. [PMID: 35266837 DOI: 10.1080/10408398.2022.2048291] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sleep disorders have received widespread attention nowadays, which have been promoted by the accelerated pace of life, unhealthy diets and lack of exercise in modern society. The chemical medications to improve sleep has shown serious side effects and risks with high costs. Therefore, it is urgent to develop efficient nutraceuticals from natural sources to ensure sleep quality as a sustainable strategy. As the second most consumed beverage worldwide, the health-promoting effects of tea have long been widely recognized. However, the modulatory effect of teas on sleep disorders has received much less attention. Tea contains various natural sleep-modulating active ingredients such as L-theanine (LTA), caffeine, tea polyphenols (TPP), tea pigments, tea polysaccharides (TPS) and γ-aminobutyric acid (GABA). This review focuses on the potential influence and main regulating mechanisms of different tea active ingredients on sleep, including being absorbed by the small intestine and then cross the blood-brain barrier to act on neurons in the brain as neurotransmitters, manipulating the immune system and further affect sleep-wake cycle by regulating the levels of cytokines, and controlling the gut microbes to maintain the homeostasis of circadian rhythm. Current research progress and limitations are summarized and several future development directions are also proposed. This review hopes to provide new insights into the future elucidation of the sleep-regulating mechanisms of different teas and their natural active ingredients and the development of tea-based functional foods for alleviating sleep disorders. HighlightsNatural sleep-modulating active ingredients in tea have been summarized.Influences of drinking tea or tea active ingredients on sleep are reviewed.Three main regulating mechanisms of tea active ingredients on sleep are explained.The associations among nervous system, immune system and intestinal microbiota are investigated.The potential of developing delivery carriers for tea active ingredients is proposed.
Collapse
Affiliation(s)
- Yang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jia Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Siwei Miao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Kang Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lanlan Peng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Yuanfeng Wang
- College of Life Sciences, Shanghai Normal University, Shanghai, P.R. China
| | - Xinlin Wei
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
24
|
Kim TH, Bormate KJ, Custodio RJP, Cheong JH, Lee BK, Kim HJ, Jung YS. Involvement of the adenosine A 1 receptor in the hypnotic effect of rosmarinic acid. Biomed Pharmacother 2022; 146:112483. [PMID: 34891112 DOI: 10.1016/j.biopha.2021.112483] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 11/18/2022] Open
Abstract
Insomnia, the most common sleep disorder, is characterized by a longer sleep latency, greater sleep fragmentation, and consequent excessive daytime fatigue. Due to the various side effects of prescribed hypnotics, demand for new drugs is still high. Recent studies have suggested the adenosine receptor (AR) as a potential therapeutic target for insomnia, however, clinically useful hypnotics targeting AR are not yet available. In the present study, we evaluated the hypnotic effect of rosmarinic acid, a phenolic compound widely found in medicinal plants, through pentobarbital-induced sleep test, electroencephalography/electromyography (EEG/EMG), and immunohistochemistry in mice. The underlying mechanisms were assessed by pharmacological approach using 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) and SCH5826, antagonists for A1R and A2AR, respectively. Receptor-binding assay and functional agonism were also performed. Our study provides a new evidence that rosmarinic acid has a direct binding activity (Ki = 14.21 ± 0.3 μM) and agonistic activity for A1R. We also found that rosmarinic acid significantly decreased sleep fragmentation and onset latency to NREM sleep, and these effects were abolished by DPCPX. The results from c-Fos immunostaining showed that rosmarinic acid decreased the neuronal activity in wake-promoting brain regions, such as the basal forebrain and the lateral hypothalamus, while increasing the neuronal activity in the ventrolateral preoptic nucleus, a sleep-promoting region; all these effects were significantly inhibited by DPCPX. Taken together, this study suggests that rosmarinic acid possesses novel activity as an A1R agonist and thereby exerts a hypnotic effect, and thus it may serve as a potential therapeutic agent for insomnia through targeting A1R.
Collapse
Affiliation(s)
- Tae-Ho Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Katrina Joy Bormate
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | | | - Jae Hoon Cheong
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Bo Kyung Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute in Neuroscience, Sahmyook University, Seoul 01795, Republic of Korea.
| | - Yi-Sook Jung
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
25
|
Van Egroo M, Koshmanova E, Vandewalle G, Jacobs HI. Importance of the locus coeruleus-norepinephrine system in sleep-wake regulation: implications for aging and Alzheimer’s disease. Sleep Med Rev 2022; 62:101592. [PMID: 35124476 PMCID: PMC9064973 DOI: 10.1016/j.smrv.2022.101592] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
Abstract
Five decades ago, seminal studies positioned the brainstem locus coeruleus (LC) norepinephrine (NE) system as a key substrate for the regulation of wakefulness and sleep, and this picture has recently been elaborated thanks to methodological advances in the precise investigation and experimental modulation of LC structure and functions. This review presents and discusses findings that support the major role of the LC-NE system at different levels of sleep-wake organization, ranging from its involvement in the overall architecture of the sleep-wake cycle to its associations with sleep microstructure, while accounting for the intricate neuroanatomy surrounding the LC. Given the particular position held by the LC-NE system by being at the intersection of sleep-wake dysregulation and initial pathophysiological processes of Alzheimer's disease (AD), we conclude by examining emerging opportunities to investigate LC-NE mediated relationships between sleep-wake alteration and AD in human aging. We further propose several research perspectives that could support the LC-NE system as a promising target for the identification of at-risk individuals in the preclinical stages of AD, and for the development of novel preventive interventions.
Collapse
|
26
|
The interplay between Sleep and Gut Microbiota. Brain Res Bull 2022; 180:131-146. [DOI: 10.1016/j.brainresbull.2021.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023]
|
27
|
Mehramiz M, Porter T, Laws SM, Rainey-Smith SR. Sleep, Sirtuin 1 and Alzheimer's disease: A review. AGING BRAIN 2022; 2:100050. [PMID: 36908890 PMCID: PMC9997138 DOI: 10.1016/j.nbas.2022.100050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Sleep plays a major role in brain health, and cognition. Disrupted sleep is a well-described symptom of Alzheimer's disease (AD). However, accumulating evidence suggests suboptimal sleep also increases AD risk. The deacetylase Sirtuin 1 (Sirt 1), encoded by the SIRT1 gene, impacts sleep via its relationship to wake-sleep neurotransmitters and somnogens. Evidence from animal and human studies supports a significant and complex relationship between sleep, Sirt 1/ SIRT1 and AD. Numerous hypotheses attempt to explain the critical impact of Sirt 1/ SIRT1 on wake- and sleep- promoting neurons, their related mechanisms and neurotransmitters. However, there is a paucity of studies assessing the interaction between sleep and Sirt 1/ SIRT1, as a principal component of sleep regulation, on AD pathology. In this review, we explore the potential association between Sirt 1/ SIRT1, sleep, and AD aetiology. Given sleep is a likely modifiable risk factor for AD, and recent studies suggest Sirt 1/ SIRT1 activation can be modulated by lifestyle or dietary approaches, further research in this area is required to explore its potential as a target for AD prevention and treatment.
Collapse
Affiliation(s)
- Mehrane Mehramiz
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.,School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.,Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, WA, Australia.,Australian Alzheimer's Research Foundation (Ralph and Patricia Sarich Neuroscience Research Institute), Nedlands, WA, Australia.,Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, WA, Australia.,School of Psychological Science, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
28
|
Abstract
A molecular circadian clock exists not only in the brain, but also in most cells of the body. Research over the past two decades has demonstrated that it directs daily rhythmicity of nearly every aspect of metabolism. It also consolidates sleep-wake behavior each day into an activity/feeding period and a sleep/fasting period. Otherwise, sleep-wake states are mostly controlled by hypothalamic and thalamic regulatory circuits in the brain that direct overall brain state. Recent evidence suggests that hypothalamic control of appetite and metabolism may be concomitant with sleep-wake regulation, and even share the same control centers. Thus, circadian control of metabolic pathways might be overlaid by sleep-wake control of the same pathways, providing a flexible and redundant system to modify metabolism according to both activity and environment.
Collapse
|
29
|
Guo R, Shi AM, Deng L, Li L, Wang LC, Oteng AB, Wei MP, Zhao ZH, Hooiveld G, Zhang C, Wang Q. Flavonoid-Like Components of Peanut Stem and Leaf Extract Promote Sleep by Decreasing Neuronal Excitability. Mol Nutr Food Res 2021; 66:e2100210. [PMID: 34747100 DOI: 10.1002/mnfr.202100210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/10/2021] [Indexed: 12/16/2022]
Abstract
SCOPE Peanut stem and leaf (PSL), a traditional Chinese medicine, is widely used as a dietary supplement to improve sleep quality; however, the underlying mechanism is unclear. Here, the study aims to determine whether active compounds in PSL extract exert their effects by mediating neuronal excitability. METHODS AND RESULTS Aqueous PSL extract (500 mg kg-1 BW) increases the duration of total sleep (TS), slow wave sleep (SWS) and rapid eye movement sleep (REMS) in BALB/c mice after 7 and 14 continuous days of intragastric administration. Two PSL extract components with flavonoid-like structures: 4',7-di-O-methylnaringenin (DMN, 61 µg kg-1 BW) and 2'-O-methylisoliquiritigenin (MIL, 12 µg kg-1 BW), show similar effects on sleep in BALB/c mice. Moreover, incubation with DMN (50 µM) and MIL (50 µM) acutely reduces voltage-gated sodium and potassium currents and suppresses the firing of evoked action potential in mouse cortical neurons, indicating the inhibition on neuronal excitability. Meanwhile, RNA-seq analysis predicts the potential regulation of voltage-gated channels, which is according with the molecular docking simulation that both MIL and DMN can bind to voltage gated sodium channels 1.2 (Nav 1.2). CONCLUSIONS DMN and MIL are the active ingredients of PSL that improve sleep quality, suggesting that PSL promotes sleep by regulating the excitability of neurons.
Collapse
Affiliation(s)
- Rui Guo
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Ai-Min Shi
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Lei Deng
- Nutrition, Metabolism and Genomics Group, Human Nutrition and Health Division, Wageningen University and Research, Wageningen, WE 6708, The Netherlands
| | - Lei Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Anhui, 230031, China
| | - Lie-Chen Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Anhui, 230031, China
| | | | - Meng-Ping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zhi-Hao Zhao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Guido Hooiveld
- Nutrition, Metabolism and Genomics Group, Human Nutrition and Health Division, Wageningen University and Research, Wageningen, WE 6708, The Netherlands
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Qiang Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| |
Collapse
|
30
|
Mo M, Tang Y, Wei L, Qiu J, Peng G, Lin Y, Zhou M, Dai W, Zhang Z, Chen X, Liu H, Ding L, Ye P, Wu Y, Zhu X, Wu Z, Guo W, Xu P. Soluble Triggering Receptor Expressed on Myeloid Cells 2 From Cerebrospinal Fluid in Sleep Disorders Related to Parkinson's Disease. Front Aging Neurosci 2021; 13:753210. [PMID: 34658845 PMCID: PMC8511683 DOI: 10.3389/fnagi.2021.753210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial receptor exclusively expressed in the central nervous system (CNS). It contributes to abnormal protein aggregation in neurodegenerative disorders, but its role in Parkinson’s disease (PD) is still unclear. Methods: In this case-control study, we measured the concentration of the soluble fragment of TREM2 (sTREM2) in PD patients, evaluated their sleep conditions by the PD sleep scale (PDSS), and analyzed the relationship between sTREM2 and PD symptoms. Results: We recruited 80 sporadic PD patients and 65 healthy controls without disease-related variants in TREM2. The concentration of sTREM2 in the CSF was significantly higher in PD patients than in healthy controls (p < 0.01). In the PD group, the concentration of sTREM2 had a positive correlation with α-syn in the CSF (Pearson r = 0.248, p = 0.027). Receiver operating characteristic curve (ROC) analyses showed that sTREM2 in the CSF had a significant diagnostic value for PD (AUC, 0.791; 95% CI, 0.711–0.871, p < 0.05). The subgroup analysis showed that PD patients with sleep disorders had a significantly higher concentration of sTREM2 in their CSF (p < 0.01). The concentration of sTREM2 in the CSF had a negative correlation with the PDSS score in PD patients (Pearson r = −0.555, p < 0.01). The ROC analyses showed that sTREM2 in the CSF had a significant diagnostic value for sleep disorders in PD (AUC, 0.733; 95% CI, 0.619–0.846, p < 0.05). Conclusion: Our findings suggest that CSF sTREM2 may be a potential biomarker for PD and it could help predict sleep disorders in PD patients, but multicenter prospective studies with more participants are still needed to confirm its diagnostic value in future.
Collapse
Affiliation(s)
- Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuting Tang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijian Wei
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Miaomiao Zhou
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Dai
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanqun Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Panghai Ye
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yijuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoqin Zhu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
31
|
Raymond JS, Rehn S, Hoyos CM, Bowen MT. The influence of oxytocin-based interventions on sleep-wake and sleep-related behaviour and neurobiology: A systematic review of preclinical and clinical studies. Neurosci Biobehav Rev 2021; 131:1005-1026. [PMID: 34673110 DOI: 10.1016/j.neubiorev.2021.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022]
Abstract
The oxytocin (OXT) system has garnered considerable interest due to its influence on diverse behaviours. However, scant research has considered the influence of oxytocin on sleep-wake and sleep-related behaviour and neurobiology. Consequently, the objective of this systematic review was to assess the extant preclinical and clinical evidence for the influence of oxytocin-based interventions on sleep-wake outcomes. The primary search was conducted on 22/7/2020 using six electronic databases; 30 studies (19 preclinical, 11 clinical) were included based on inclusion criteria. Studies were evaluated for risk of bias using the SYRCLE tool and the Cochrane risk of bias tools for preclinical and clinical studies, respectively. Results indicated manipulation of the OXT system can influence sleep-wake outcomes. Preclinical evidence suggests a wake-promoting influence of OXT system activation whereas the clinical evidence suggests little or no sleep-promoting influence of OXT. OXT dose was identified as a likely modulatory factor of OXT-induced effects on sleep-wake behaviour. Future studies are necessary to validate and strengthen these tentative conclusions about the influence of OXT on sleep-wake behaviour.
Collapse
Affiliation(s)
- Joel S Raymond
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia
| | - Simone Rehn
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia
| | - Camilla M Hoyos
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia; The University of Sydney, Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Camperdown, NSW, Australia
| | - Michael T Bowen
- The University of Sydney, Faculty of Science, School of Psychology, Camperdown, NSW, Australia; The University of Sydney, Brain and Mind Centre, Camperdown, NSW, Australia.
| |
Collapse
|
32
|
Van Egroo M, van Hooren RWE, Jacobs HIL. Associations between locus coeruleus integrity and nocturnal awakenings in the context of Alzheimer's disease plasma biomarkers: a 7T MRI study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:159. [PMID: 34560904 PMCID: PMC8464124 DOI: 10.1186/s13195-021-00902-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/12/2021] [Indexed: 12/12/2022]
Abstract
Background The brainstem locus coeruleus (LC) constitutes the intersection of the initial pathophysiological processes of Alzheimer’s disease (AD) and sleep-wake dysregulation in the preclinical stages of the disease. However, the interplay between in vivo assessment of LC degeneration and AD-related sleep alterations remains unknown. Here, we sought to investigate whether MRI-assessed LC structural integrity relates to subjective sleep-wake measures in the context of AD plasma biomarkers, in cognitively unimpaired older individuals. Methods Seventy-two cognitively unimpaired older individuals aged 50–85 years (mean age = 65.2 ± 8.2 years, 37 women, 21 APOE ε4 carriers) underwent high-resolution imaging of the LC at 7 Tesla, and LC structural integrity was quantified using a data-driven approach. Reports on habitual sleep quality and nocturnal awakenings were collected using sleep questionnaires. Plasma levels of total tau, p-tau181, Aβ40, and Aβ42 were measured using single-molecule array technology. Results Intensity-based cluster analyses indicated two distinct LC segments, with one covering the middle-to-caudal LC and displaying lower intensity compared to the middle-to-rostral cluster (t70 = −5.12, p < 0.0001). After correction for age, sex, depression, and APOE status, lower MRI signal intensity within the middle-to-caudal LC was associated with a higher number of self-reported nocturnal awakenings (F1,63 = 6.73, pFDR = 0.03). Furthermore, this association was mostly evident in individuals with elevated levels of total tau in the plasma (F1,61 = 4.26, p = 0.04). Conclusion Our findings provide in vivo evidence that worse LC structural integrity is associated with more frequent nocturnal awakenings in the context of neurodegeneration, in cognitively unimpaired older individuals. These results support the critical role of the LC for sleep-wake regulation in the preclinical stages of AD and hold promises for the identification of at-risk populations for preventive interventions. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00902-8.
Collapse
Affiliation(s)
- Maxime Van Egroo
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, UNS40 box 34, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Roy W E van Hooren
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, UNS40 box 34, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Heidi I L Jacobs
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, UNS40 box 34, P.O. Box 616, 6200 MD, Maastricht, The Netherlands. .,Faculty of Psychology and Neuroscience, Department of Cognitive Neuroscience, Maastricht University, Maastricht, The Netherlands. .,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
34
|
Tau-driven degeneration of sleep- and wake-regulating neurons in Alzheimer's disease. Sleep Med Rev 2021; 60:101541. [PMID: 34500400 DOI: 10.1016/j.smrv.2021.101541] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/22/2021] [Accepted: 08/06/2021] [Indexed: 11/22/2022]
Abstract
Disturbances of the sleep/wake cycle in Alzheimer's disease (AD) are common, frequently precede cognitive decline, and tend to worsen with disease progression. Sleep is critical to the maintenance of homeostatic and circadian function, and chronic sleep disturbances have significant cognitive and physical health consequences that likely exacerbate disease severity. Sleep-wake cycles are regulated by neuromodulatory centers located in the brainstem, the hypothalamus, and the basal forebrain, many of which are vulnerable to the accumulation of abnormal protein deposits associated with neurodegenerative conditions. In AD, while sleep disturbances are commonly attributed to the accumulation of amyloid beta, patients often first experience sleep issues prior to the appearance of amyloid beta plaques, on a timeline that more closely corresponds to the first appearance of abnormal tau neurofibrillary tangles in sleep/wake regulating areas of the brainstem. Sleep disturbances also occur in pure tauopathies, providing further support that tau is a major contributor. Here, we provide an overview of the neuroanatomy of sleep/wake centers discovered in animal models, and review the evidence that tau-driven neuropathology is a primary driver of sleep disturbance in AD.
Collapse
|
35
|
Altenhofen S, Bonan CD. Zebrafish as a tool in the study of sleep and memory-related disorders. Curr Neuropharmacol 2021; 20:540-549. [PMID: 34254919 DOI: 10.2174/1570159x19666210712141041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/23/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
Sleep is an evolutionarily conserved phenomenon, being an essential biological necessity for the learning process and memory consolidation. The brain displays two types of electrical activity during sleep: slow-wave activity or non-rapid eye movement (NREM) sleep and desynchronized brain wave activity or rapid eye movement (REM) sleep. There are many theories about "Why we need to sleep?" among them the synaptic homeostasis. This theory proposes that the role of sleep is the restoration of synaptic homeostasis, which is destabilized by synaptic strengthening triggered by learning during waking and by synaptogenesis during development. Sleep diminishes the plasticity load on neurons and other cells to normalize synaptic strength. In contrast, it re-establishes neuronal selectivity and the ability to learn, leading to the consolidation and integration of memories. The use of zebrafish as a tool to assess sleep and its disorders is growing, although sleep in this animal is not yet divided, for example, into REM and NREM states. However, zebrafish are known to have a regulated daytime circadian rhythm. Their sleep state is characterized by periods of inactivity accompanied by an increase in arousal threshold, preference for resting place, and the "rebound sleep effect" phenomenon, which causes an increased slow-wave activity after a forced waking period. In addition, drugs known to modulate sleep, such as melatonin, nootropics, and nicotine, have been tested in zebrafish. In this review, we discuss the use of zebrafish as a model to investigate sleep mechanisms and their regulation, demonstrating this species as a promising model for sleep research.
Collapse
Affiliation(s)
- Stefani Altenhofen
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celulare Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celulare Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, porto Alegre, RS, Brazil
| |
Collapse
|
36
|
Galeb HA, Wilkinson EL, Stowell AF, Lin H, Murphy ST, Martin‐Hirsch PL, Mort RL, Taylor AM, Hardy JG. Melanins as Sustainable Resources for Advanced Biotechnological Applications. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000102. [PMID: 33552556 PMCID: PMC7857133 DOI: 10.1002/gch2.202000102] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/04/2020] [Indexed: 05/17/2023]
Abstract
Melanins are a class of biopolymers that are widespread in nature and have diverse origins, chemical compositions, and functions. Their chemical, electrical, optical, and paramagnetic properties offer opportunities for applications in materials science, particularly for medical and technical uses. This review focuses on the application of analytical techniques to study melanins in multidisciplinary contexts with a view to their use as sustainable resources for advanced biotechnological applications, and how these may facilitate the achievement of the United Nations Sustainable Development Goals.
Collapse
Affiliation(s)
- Hanaa A. Galeb
- Department of ChemistryLancaster UniversityLancasterLA1 4YBUK
- Department of ChemistryScience and Arts CollegeRabigh CampusKing Abdulaziz UniversityJeddah21577Saudi Arabia
| | - Emma L. Wilkinson
- Department of Biomedical and Life SciencesLancaster UniversityLancasterLA1 4YGUK
| | - Alison F. Stowell
- Department of Organisation, Work and TechnologyLancaster University Management SchoolLancaster UniversityLancasterLA1 4YXUK
| | - Hungyen Lin
- Department of EngineeringLancaster UniversityLancasterLA1 4YWUK
| | - Samuel T. Murphy
- Department of EngineeringLancaster UniversityLancasterLA1 4YWUK
- Materials Science InstituteLancaster UniversityLancasterLA1 4YBUK
| | - Pierre L. Martin‐Hirsch
- Lancashire Teaching Hospitals NHS TrustRoyal Preston HospitalSharoe Green LanePrestonPR2 9HTUK
| | - Richard L. Mort
- Department of Biomedical and Life SciencesLancaster UniversityLancasterLA1 4YGUK
| | - Adam M. Taylor
- Lancaster Medical SchoolLancaster UniversityLancasterLA1 4YWUK
| | - John G. Hardy
- Department of ChemistryLancaster UniversityLancasterLA1 4YBUK
- Materials Science InstituteLancaster UniversityLancasterLA1 4YBUK
| |
Collapse
|
37
|
Falup-Pecurariu C, Diaconu Ș, Țînț D, Falup-Pecurariu O. Neurobiology of sleep (Review). Exp Ther Med 2021; 21:272. [PMID: 33603879 DOI: 10.3892/etm.2021.9703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Sleep is a physiological global state composed of two different phases: Non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. The control mechanisms of sleep manifest at the level of genetic, biological and cellular organization. Several brain areas, including the basal forebrain, thalamus, and hypothalamus, take part in regulating the activity of this status of life. The signals between different brain regions and those from cortical areas to periphery are conducted through various neuromediators, which are known to either promote wakefulness or sleep. Among others, serotonin, norepinephrine, histamine, hypocretin (orexin), acetylcholine, dopamine, glutamate, and gamma-aminobutyric acid are known to orchestrate the intrinsic mechanisms of sleep neurobiology. Several models that explain the transition and the continuity between wakefulness, NREM sleep and REM sleep have been proposed. All of these models include neurotransmitters as ligands in a complex reciprocal connectivity across the key-centers taking part in the regulation of sleep. Moreover, various environmental cues are integrated by a central pacemaker-located in the suprachiasmatic nucleus-which is able to connect with cortical regions and with peripheral tissues in order to promote the sleep-wake pattern.
Collapse
Affiliation(s)
- Cristian Falup-Pecurariu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Department of Neurology, County Emergency Clinic Hospital, 500365 Brașov, Romania
| | - Ștefania Diaconu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Department of Neurology, County Emergency Clinic Hospital, 500365 Brașov, Romania
| | - Diana Țînț
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Clinicco Hospital, 500059 Brașov, Romania
| | - Oana Falup-Pecurariu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania
| |
Collapse
|
38
|
Zhao FY, Fu QQ, Spencer SJ, Kennedy GA, Conduit R, Zhang WJ, Zheng Z. Acupuncture: A Promising Approach for Comorbid Depression and Insomnia in Perimenopause. Nat Sci Sleep 2021; 13:1823-1863. [PMID: 34675729 PMCID: PMC8520448 DOI: 10.2147/nss.s332474] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/24/2021] [Indexed: 12/12/2022] Open
Abstract
Comorbid depression and insomnia are ubiquitous mental complaints among women going through the perimenopausal stage of life and can result in major decline in quality of life. Antidepressive agents combined with/without hypnotics, and/or hormone therapy are currently the most common treatment for perimenopausal depression (PMD) and insomnia (PMI). Balancing the benefits of these pharmacotherapies against the risk of adverse events (AEs) is a difficult task for both clinicians and women. There has been a growing body of research regarding the utilization of acupuncture for treatment of PMD or PMI, whereas no studies of acupuncture for comorbid PMD and PMI have appeared. In this review, we summarize the clinical and preclinical evidence of acupuncture as a treatment for PMD or PMI, and then discuss the potential mechanisms involved and the role of acupuncture in helping women during this transition. Most clinical trials indicate that acupuncture ameliorates not only PMD/PMI but also climacteric symptoms with minimal AEs. It also regulates serum hormone levels. The reliability of trials is however limited due to methodological flaws in most studies. Rodent studies suggest that acupuncture prolongs total sleep time and reduces depression-like behavior in PMI and PMD models, respectively. These effects are possibly mediated through multiple mechanisms of action, including modulating sex hormones, neurotransmitters, hypothalamic-pituitary-adrenal axis/hypothalamic-pituitary-ovary axis, oxidative stress, signaling pathways, and other cellular events. In conclusion, acupuncture is a promising therapeutic strategy for comorbid depression and insomnia during perimenopause. Neuroendocrine modulation is likely to play a major role in mediating those effects. High-quality trials are required to further validate acupuncture's effectiveness.
Collapse
Affiliation(s)
- Fei-Yi Zhao
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia.,Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, People's Republic of China.,Department of Nursing, School of International Medical Technology, Shanghai Sanda University, Shanghai, 201209, People's Republic of China
| | - Qiang-Qiang Fu
- Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, People's Republic of China
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia.,ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Gerard A Kennedy
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia.,School of Science, Psychology and Sport, Federation University, Mount Helen, Victoria, Australia.,Institute for Breathing and Sleep, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Russell Conduit
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Wen-Jing Zhang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, People's Republic of China
| | - Zhen Zheng
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| |
Collapse
|
39
|
Harris SS, Schwerd-Kleine T, Lee BI, Busche MA. The Reciprocal Interaction Between Sleep and Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1344:169-188. [PMID: 34773232 DOI: 10.1007/978-3-030-81147-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
It is becoming increasingly recognized that patients with a variety of neurodegenerative diseases exhibit disordered sleep/wake patterns. While sleep impairments have typically been thought of as sequelae of underlying neurodegenerative processes in sleep-wake cycle regulating brain regions, including the brainstem, hypothalamus, and basal forebrain, emerging evidence now indicates that sleep deficits may also act as pathophysiological drivers of brain-wide disease progression. Specifically, recent work has indicated that impaired sleep can impact on neuronal activity, brain clearance mechanisms, pathological build-up of proteins, and inflammation. Altered sleep patterns may therefore be novel (potentially reversible) dynamic functional markers of proteinopathies and modifiable targets for early therapeutic intervention using non-invasive stimulation and behavioral techniques. Here we highlight research describing a potentially reciprocal interaction between impaired sleep and circadian patterns and the accumulation of pathological signs and features in Alzheimer's disease, the most prevalent neurodegenerative disease in the elderly.
Collapse
Affiliation(s)
| | | | - Byung Il Lee
- UK Dementia Research Institute at UCL, London, UK
| | | |
Collapse
|
40
|
Richards A, Kanady JC, Neylan TC. Sleep disturbance in PTSD and other anxiety-related disorders: an updated review of clinical features, physiological characteristics, and psychological and neurobiological mechanisms. Neuropsychopharmacology 2020; 45:55-73. [PMID: 31443103 PMCID: PMC6879567 DOI: 10.1038/s41386-019-0486-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 02/08/2023]
Abstract
The current report provides an updated review of sleep disturbance in posttraumatic stress disorder and anxiety-related disorders. First, this review provides a summary description of the unique and overlapping clinical characteristics and physiological features of sleep disturbance in specific DSM anxiety-related disorders. Second, this review presents evidence of a bidirectional relationship between sleep disturbance and anxiety-related disorders, and provides a model to explain this relationship by integrating research on psychological and neurocognitive processes with a current understanding of neurobiological pathways. A heuristic neurobiological framework for understanding the bidirectional relationship between abnormalities in sleep and anxiety-related brain pathways is presented. Directions for future research are suggested.
Collapse
Affiliation(s)
- Anne Richards
- The San Francisco VA Health Care System, San Francisco, CA, USA.
- The University of California, San Francisco, San Francisco, CA, USA.
| | - Jennifer C Kanady
- The San Francisco VA Health Care System, San Francisco, CA, USA
- The University of California, San Francisco, San Francisco, CA, USA
| | - Thomas C Neylan
- The San Francisco VA Health Care System, San Francisco, CA, USA
- The University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
41
|
Oh J, Eser RA, Ehrenberg AJ, Morales D, Petersen C, Kudlacek J, Dunlop SR, Theofilas P, Resende EDPF, Cosme C, Alho EJL, Spina S, Walsh CM, Miller BL, Seeley WW, Bittencourt JC, Neylan TC, Heinsen H, Grinberg LT. Profound degeneration of wake-promoting neurons in Alzheimer's disease. Alzheimers Dement 2019; 15:1253-1263. [PMID: 31416793 DOI: 10.1016/j.jalz.2019.06.3916] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/10/2019] [Accepted: 06/10/2019] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Sleep-wake disturbances are a common and early feature in Alzheimer's disease (AD). The impact of early tau pathology in wake-promoting neurons (WPNs) remains unclear. METHODS We performed stereology in postmortem brains from AD individuals and healthy controls to identify quantitative differences in morphological metrics in WPNs. Progressive supranuclear palsy (PSP) and corticobasal degeneration were included as disease-specific controls. RESULTS The three nuclei studied accumulate considerable amounts of tau inclusions and showed a decrease in neurotransmitter-synthetizing neurons in AD, PSP, and corticobasal degeneration. However, substantial neuronal loss was exclusively found in AD. DISCUSSION WPNs are extremely vulnerable to AD but not to 4 repeat tauopathies. Considering that WPNs are involved early in AD, such degeneration should be included in the models explaining sleep-wake disturbances in AD and considered when designing a clinical intervention. Sparing of WPNs in PSP, a condition featuring hyperinsomnia, suggest that interventions to suppress the arousal system may benefit patients with PSP.
Collapse
Affiliation(s)
- Jun Oh
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Rana A Eser
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Dulce Morales
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Cathrine Petersen
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jessica Kudlacek
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sara R Dunlop
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Panos Theofilas
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Elisa D P F Resende
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Celica Cosme
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Eduardo J L Alho
- Department of Neurology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Salvatore Spina
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Christine M Walsh
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jackson C Bittencourt
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Center for Neuroscience and Behavior, Institute of Psychology, University of Sao Paulo, Sao Paulo, Brazil
| | - Thomas C Neylan
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Helmut Heinsen
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Psychiatry, University of Wuerzburg, Wuerzburg, Germany
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Global Brain Health Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
42
|
Chazot PL, Johnston L, Mcauley E, Bonner S. Histamine and Delirium: Current Opinion. Front Pharmacol 2019; 10:299. [PMID: 31024298 PMCID: PMC6467166 DOI: 10.3389/fphar.2019.00299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
Delirium is a very common, but refractory clinical state, notably present in intensive care and in the growing aging community. It is characterized by fluctuating disturbances in a number of key behavioral features, namely cognition, mood, attention, arousal, and self-awareness. Histamine is arguably the most pleotropic neurotransmitter in the human brain, and this review provides a rationale, and proposes that this neuroactive amine plays a role in modulating the characteristic features of delirium. While centrally permeable H1 and H2 histamine receptor antagonists have pro-delirium potential, we propose that centrally permeable H3 histamine receptor antagonists may provide an exciting new strategy to combat delirium. The Histamine H4 receptor may also have an indirect inflammatory neuroglial role which requires further exploration.
Collapse
Affiliation(s)
- Paul L. Chazot
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Laura Johnston
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Edel Mcauley
- Intensive Care, South Tees Hospitals NHS Foundation Trust, The James Cook University Hospital, Middlesbrough, United Kingdom
| | - Stephen Bonner
- Intensive Care, South Tees Hospitals NHS Foundation Trust, The James Cook University Hospital, Middlesbrough, United Kingdom
| |
Collapse
|