1
|
Chen ZK, Liu YY, Zhou JC, Chen GH, Liu CF, Qu WM, Huang ZL. Insomnia-related rodent models in drug discovery. Acta Pharmacol Sin 2024; 45:1777-1792. [PMID: 38671193 PMCID: PMC11335876 DOI: 10.1038/s41401-024-01269-w] [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: 11/09/2023] [Accepted: 03/24/2024] [Indexed: 04/28/2024]
Abstract
Despite the widespread prevalence and important medical impact of insomnia, effective agents with few side effects are lacking in clinics. This is most likely due to relatively poor understanding of the etiology and pathophysiology of insomnia, and the lack of appropriate animal models for screening new compounds. As the main homeostatic, circadian, and neurochemical modulations of sleep remain essentially similar between humans and rodents, rodent models are often used to elucidate the mechanisms of insomnia and to develop novel therapeutic targets. In this article, we focus on several rodent models of insomnia induced by stress, diseases, drugs, disruption of the circadian clock, and other means such as genetic manipulation of specific neuronal activity, respectively, which could be used to screen for novel hypnotics. Moreover, important advantages and constraints of some animal models are discussed. Finally, this review highlights that the rodent models of insomnia may play a crucial role in novel drug development to optimize the management of insomnia.
Collapse
Affiliation(s)
- Ze-Ka Chen
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yuan-Yuan Liu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ji-Chuan Zhou
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science; Joint International Research Laboratory of Sleep; and Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Li Q, Bai J, Ma Y, Sun Y, Zhou W, Wang Z, Zhou Z, Wang Z, Chen Y, Abliz Z. Pharmacometabolomics and mass spectrometry imaging approach to reveal the neurochemical mechanisms of Polygala tenuifolia. J Pharm Anal 2024; 14:100973. [PMID: 39175609 PMCID: PMC11340588 DOI: 10.1016/j.jpha.2024.100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/02/2024] [Accepted: 03/25/2024] [Indexed: 08/24/2024] Open
Abstract
Polygala tenuifolia, commonly known as Yuanzhi (YZ) in Chinese, has been shown to possess anti-insomnia properties. However, the material basis and the mechanism underlying its sedative-hypnotic effects remain unclear. Herein, we investigated the active components and neurochemical mechanism of YZ extracts using liquid chromatography tandem mass spectrometry (LC-MS/MS)-based pharmacometabolomics and mass spectrometry imaging (MSI)-based spatial resolved metabolomics. According to the results, 17 prototypes out of 101 ingredients in the YZ extract were detected in both the plasma and brain, which might be the major components contributing to the sedative-hypnotic effects. Network pharmacology analysis revealed that these prototypes may exert their effects through neuroactive ligand-receptor interaction, serotonergic synapse, dopaminergic synapse, and dopaminergic synapse, among other pathways. LC-MS/MS-based targeted metabolomics and Western blot (WB) revealed that tryptophan-serotonin-melatonin (Trp-5-HT-Mel) and tyrosine-norepinephrine-adrenaline (Tyr-Ne-Ad) are the key regulated pathways. Dopa decarboxylase (DDC) upregulation and phenylethanolamine N-methyltransferase (PNMT) downregulation further confirmed these pathways. Furthermore, MSI-based spatially resolved metabolomics revealed notable alterations in 5-HT in the pineal gland (PG), and Ad in the brainstem, including the middle brain (MB), pons (PN), and hypothalamus (HY). In summary, this study illustrates the efficacy of an integrated multidimensional metabolomics approach in unraveling the sedative-hypnotic effects and neurochemical mechanisms of a Chinese herbal medicine, YZ.
Collapse
Affiliation(s)
- Qian Li
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Jinpeng Bai
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yuxue Ma
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Yu Sun
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Wenbin Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Zhaoying Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Zhi Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Zhonghua Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yanhua Chen
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Zeper Abliz
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (National Ethnic Affairs Commission), Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
3
|
Cirrincione L, Plescia F, Malta G, Campagna M, Lecca LI, Skerjanc A, Carena E, Baylon V, Theodoridou K, Fruscione S, Cannizzaro E. Evaluation of Correlation between Sleep and Psychiatric Disorders in a Population of Night Shift Workers: A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3756. [PMID: 36834452 PMCID: PMC9967097 DOI: 10.3390/ijerph20043756] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Insomnia is the perception of inadequate, insufficient or non-restorative sleep. Of all sleep-related disorders, insomnia is the most common. It is important to remember that the sleep-wake cycle also plays a central role in the genesis of anxiety and depression. The aim of our study is to evaluate the association between sleep disturbances and anxiety and depression in a group of workers of both sexes who perform night shift work. METHODS Information on sleep disorders was collected by administering the Insomnia Severity Index (ISI) questionnaire. Statistical analysis was conducted using the Chi-square test to assess whether there were any differences between sex for those who were healthy or who were diagnosed with psychiatric disorders. RESULTS The results showed that there was a good percentage of subjects with insomnia problems, impairing normal daily activities and promoting the onset of fatigue, daytime sleepiness, cognitive performance deficits and mood disorders. CONCLUSION We highlighted how anxious and depressive anxiety disorders are more pronounced in people who suffer from altered sleep-wake rhythms. Further research in this direction could prove to be fundamental for understanding the genesis of the onset of other disorders as well.
Collapse
Affiliation(s)
- Luigi Cirrincione
- Department of Health Promotion Sciences Maternal and Child Care, Internal Medicine and Medical Specialties ‘Giuseppe D’Alessandro’, University of Palermo, Via del Vespro 133, 90127 Palermo, Italy
| | - Fulvio Plescia
- Department of Health Promotion Sciences Maternal and Child Care, Internal Medicine and Medical Specialties ‘Giuseppe D’Alessandro’, University of Palermo, Via del Vespro 133, 90127 Palermo, Italy
| | - Ginevra Malta
- Department of Health Promotion Sciences Maternal and Child Care, Internal Medicine and Medical Specialties ‘Giuseppe D’Alessandro’, University of Palermo, Via del Vespro 133, 90127 Palermo, Italy
| | - Marcello Campagna
- Department of Medical Sciences and Public Health, University of Cagliari, 09127 Cagliari, Italy
| | - Luigi Isaia Lecca
- Department of Medical Sciences and Public Health, University of Cagliari, 09127 Cagliari, Italy
| | - Alenka Skerjanc
- Clinical Institute for Occupational, Traffic and Sports Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Elisa Carena
- Department of Sciences of Public Health and Pediatrics, University of Turin, 10126 Turin, Italy
| | - Vincenzo Baylon
- Newton Lewis Institute Scientific Research-Life Science Park, 3000 San Gwann, Malta
| | - Kelly Theodoridou
- Department of Microbiology, Andreas Syggros University Hospital Athens Greece, 10552 Athens, Greece
| | - Santo Fruscione
- Department of Health Promotion Sciences Maternal and Child Care, Internal Medicine and Medical Specialties ‘Giuseppe D’Alessandro’, University of Palermo, Via del Vespro 133, 90127 Palermo, Italy
| | - Emanuele Cannizzaro
- Department of Health Promotion Sciences Maternal and Child Care, Internal Medicine and Medical Specialties ‘Giuseppe D’Alessandro’, University of Palermo, Via del Vespro 133, 90127 Palermo, Italy
| |
Collapse
|
4
|
Lin Y, Roy K, Ioka S, Otani R, Amezawa M, Ishikawa Y, Cherasse Y, Kaushik MK, Klewe-Nebenius D, Zhou L, Yanagisawa M, Oishi Y, Saitoh T, Lazarus M. Positive allosteric adenosine A 2A receptor modulation suppresses insomnia associated with mania- and schizophrenia-like behaviors in mice. Front Pharmacol 2023; 14:1138666. [PMID: 37153764 PMCID: PMC10155833 DOI: 10.3389/fphar.2023.1138666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Background: Insomnia is associated with psychiatric illnesses such as bipolar disorder or schizophrenia. Treating insomnia improves psychotic symptoms severity, quality of life, and functional outcomes. Patients with psychiatric disorders are often dissatisfied with the available therapeutic options for their insomnia. In contrast, positive allosteric modulation of adenosine A2A receptors (A2ARs) leads to slow-wave sleep without cardiovascular side effects in contrast to A2AR agonists. Methods: We investigated the hypnotic effects of A2AR positive allosteric modulators (PAMs) in mice with mania-like behavior produced by ablating GABAergic neurons in the ventral medial midbrain/pons area and in a mouse model of schizophrenia by knocking out of microtubule-associated protein 6. We also compared the properties of sleep induced by A2AR PAMs in mice with mania-like behavior with those induced by DORA-22, a dual orexin receptor antagonist that improves sleep in pre-clinical models, and the benzodiazepine diazepam. Results: A2AR PAMs suppress insomnia associated with mania- or schizophrenia-like behaviors in mice. A2AR PAM-mediated suppression of insomnia in mice with mania-like behavior was similar to that mediated by DORA-22, and, unlike diazepam, did not result in abnormal sleep. Conclusion: A2AR allosteric modulation may represent a new therapeutic avenue for sleep disruption associated with bipolar disorder or psychosis.
Collapse
Affiliation(s)
- Yang Lin
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Koustav Roy
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Shuji Ioka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Rintaro Otani
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Mao Amezawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yukiko Ishikawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Mahesh K. Kaushik
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Daniela Klewe-Nebenius
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Li Zhou
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- *Correspondence: Tsuyoshi Saitoh, ; Michael Lazarus,
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Ibaraki, Japan
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- *Correspondence: Tsuyoshi Saitoh, ; Michael Lazarus,
| |
Collapse
|
5
|
Dujardin S, Pijpers A, Pevernagie D. Prescription Drugs Used in Insomnia. Sleep Med Clin 2022; 17:315-328. [PMID: 36150797 DOI: 10.1016/j.jsmc.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In insomnia, the subjective aspects of the sleep complaint are paramount in the diagnostic criteria. Epidemiologic studies increasingly point to a link between insomnia and somatic morbidity and mortality, but until now, only in the subgroup of objectively poor sleepers. Although pharmacologic treatment might offer some benefits to this subgroup of insomnia patients, to date, there is no evidence that hypnotics can ameliorate their health risks. Further unraveling of the neurobiology and genetics of sleep regulation and the pathophysiology of insomnia will help the development of drugs that not only improve subjective sleep complaints but also objective health outcomes.
Collapse
Affiliation(s)
- Sylvie Dujardin
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, the Netherlands
| | - Angelique Pijpers
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, the Netherlands
| | - Dirk Pevernagie
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, the Netherlands; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, Ghent 9000, Belgium.
| |
Collapse
|
6
|
Kim H, Park I, Park K, Park S, Kim YI, Park BG. The Positive Effects of Poria cocos Extract on Quality of Sleep in Insomnia Rat Models. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116629. [PMID: 35682214 PMCID: PMC9180690 DOI: 10.3390/ijerph19116629] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
Sleep disorders may have various causes and can incur mental and/or physical symptoms, and affect an individual’s quality of life. In this study, we confirm that the Poria cocos extract (PCET) can improve sleep quality and structure by promoting inhibitory neurotransmission via the γ-aminobutyric acid (GABA) type A (GABAA) receptors based on the mechanisms revealed in the experiment with superior cervical ganglion neurons. Pentobarbital-induced sleep tests were conducted in order to determine whether the PCET extract improves the sleep quality and structure in normal ICR mice. Sleep latency and duration were checked with the righting reflex. To simulate the state of awakening as well as a normal sleep state, caffeine was administered orally before the PCET diet. After oral gavage of PCET, sleep latency was decreased, and total sleep duration was increased in normal and caffeine-induced sleep disturbance state. In the ACTH-induced sleep disturbed models, administration of PCET significantly reduced the sleep latency and increased the non-REM sleep duration, which was analyzed in real-time EEG by implanting wireless electrodes in SD rats. PCET was found to improve the sleep quality under a normal sleep state through the GABAA receptor; it also promoted and improved the sleep quality and sleep structure in both the arousal activation state and stress-based sleep disturbance.
Collapse
Affiliation(s)
- Hyeyun Kim
- The Convergence Institute of Healthcare and Medical Science, Department of Neurology, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 25601, Korea; (H.K.); (Y.I.K.)
| | - Injune Park
- Department of Emergency Medicine, Gangneung Asan Hospital, Ulsan University, Gangneung 44610, Korea;
| | - Kyunyong Park
- Department of Physiology, College of Medicine, Catholic Kwandong University, Gangneung 25601, Korea; (K.P.); (S.P.)
| | - Seohyun Park
- Department of Physiology, College of Medicine, Catholic Kwandong University, Gangneung 25601, Korea; (K.P.); (S.P.)
| | - Yeong In Kim
- The Convergence Institute of Healthcare and Medical Science, Department of Neurology, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 25601, Korea; (H.K.); (Y.I.K.)
| | - Byong-Gon Park
- Department of Physiology, College of Medicine, Catholic Kwandong University, Gangneung 25601, Korea; (K.P.); (S.P.)
- Correspondence: ; Tel.: +82-33-649-7471
| |
Collapse
|
7
|
Yang Y, Long K, Wang Y, Li L, Shi J, Liu J, Kong L, Yu L, Ding J, Huang Z, Wang W, Zhan C. NIR Light-Triggered Quantitative Pulsed Drug Release. Adv Healthc Mater 2022; 11:e2102362. [PMID: 34851048 DOI: 10.1002/adhm.202102362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Indexed: 12/12/2022]
Abstract
Quantitative drug release is important for improving therapeutic efficiency and avoiding side effects. While using long-term delivery system for repeated therapies, it is indispensable but challenging to accurately control the drug dosing. Here, a photocleavable prodrug loaded hydrogel is proposed for near infrared (NIR) light-triggered quantitative pulsed drug release. IR783, a commercially available NIR fluorescent dye, is conjugated with methyl honokiol (mHNK) to give a photocleavable IR783-mHNK prodrug. Injectable glycol chitosan (GC) hydrogel is chosen as a reservoir, in which IR783-mHNK can be efficiently loaded via electrostatic and hydrophobic interactions. Upon 680 nm light-emitting diode (LED) light irradiation, IR783-mHNK cleaves and mHNK is released. Notably, it is found that IR783-mHNK presents synchronous photocleavage-fluorescence bleaching phenomenon. The released amount of mHNK is visible by measuring the residual fluorescent intensity of hydrogel. Quantitative drug release is achieved by controlling irradiation duration and the drug release process is visible by fluorescence imaging. The prodrug-loaded hydrogel shows good stability, minimum leakage and efficient light responsibility both in vitro and in vivo. After light triggering, monitorable quantitative mHNK release and on-demand sleep-promotiing effect are verified in mice without toxicities.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacology School of Basic Medical Sciences & Center of Medical Research and Innovation Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200032 China
| | - Kaiqi Long
- State Key Laboratory of Pharmaceutical Biotechnology Department of Pharmacology and Pharmacy and Laboratory of Molecular Engineering and Nanomedicine Dr. Li Dak‐Sum Research Centre Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yiqun Wang
- Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science Fudan University Shanghai 200032 China
| | - Lei Li
- Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science Fudan University Shanghai 200032 China
| | - Jiayue Shi
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Jican Liu
- Department of Pathology Affiliated Zhongshan Hospital Qingpu Branch Fudan University Shanghai 201700 PR China
| | - Lingxi Kong
- Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science Fudan University Shanghai 200032 China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers Department of Macromolecular Science Fudan University Shanghai 200438 China
| | - Zhili Huang
- Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science Fudan University Shanghai 200032 China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology Department of Pharmacology and Pharmacy and Laboratory of Molecular Engineering and Nanomedicine Dr. Li Dak‐Sum Research Centre Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Changyou Zhan
- Department of Pharmacology School of Basic Medical Sciences & Center of Medical Research and Innovation Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers Fudan University Shanghai 200032 China
| |
Collapse
|
8
|
Belelli D, Phillips GD, Atack JR, Lambert JJ. Relating neurosteroid modulation of inhibitory neurotransmission to behaviour. J Neuroendocrinol 2022; 34:e13045. [PMID: 34644812 DOI: 10.1111/jne.13045] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/24/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022]
Abstract
Studies in the 1980s revealed endogenous metabolites of progesterone and deoxycorticosterone to be potent, efficacious, positive allosteric modulators (PAMs) of the GABAA receptor (GABAA R). The discovery that such steroids are locally synthesised in the central nervous system (CNS) promoted the thesis that neural inhibition in the CNS may be "fine-tuned" by these neurosteroids to influence behaviour. In preclinical studies, these neurosteroids exhibited anxiolytic, anticonvulsant, analgesic and sedative properties and, at relatively high doses, induced a state of general anaesthesia, a profile consistent with their interaction with GABAA Rs. However, realising the therapeutic potential of either endogenous neurosteroids or synthetic "neuroactive" steroids has proven challenging. Recent approval by the Food and Drug Administration of the use of allopregnanolone (brexanolone) to treat postpartum depression has rekindled enthusiasm for exploring their potential as new medicines. Although neurosteroids are selective for GABAA Rs, they exhibit little or no selectivity across the many GABAA R subtypes. Nevertheless, a relatively minor population of receptors incorporating the δ-subunit (δ-GABAA Rs) appears to be an important contributor to their behavioural effects. Here, we consider how neurosteroids acting upon GABAA Rs influence neuronal signalling, as well as how such effects may acutely and persistently influence behaviour, and explore the case for developing selective PAMs of δ-GABAA R subtypes for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Delia Belelli
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Grant D Phillips
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - John R Atack
- Medicines Discovery Institute, Cardiff University, Cardiff, UK
| | - Jeremy J Lambert
- Neuroscience, Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
9
|
Qian J, Zheng L, Su G, Huang M, Luo D, Zhao M. Identification and Screening of Potential Bioactive Peptides with Sleep-Enhancing Effects in Bovine Milk Casein Hydrolysate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11246-11258. [PMID: 34543014 DOI: 10.1021/acs.jafc.1c03937] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Casein tryptic hydrolysate (CTH) has been proven to possess stress-relieving and sleep-enhancing effects, but only one decapeptide YLGYLEQLLR (α-CZP) in CTH was reported to exhibit affinity for the benzodiazepine site of a GABAA receptor (GABAAR). This study aimed to compare the sleep-enhancing effects between CTH and α-CZP and to explore novel sleep-enhancing peptides. Our results showed that CTH significantly prolonged sleep duration in mice, which was almost 2-fold longer than that of α-CZP. The α-CZP in CTH was degraded more slowly than the synthetic α-CZP; meanwhile, CTH could release other potential sleep-enhancing peptides during gastrointestinal digestion. Additionally, two peptides YPVEPF and YFYPEL with strong sleep-enhancing activity were explored by virtual screening. Especially, YPVEPF could significantly prolong the sleep duration from 559.00 ± 272.24 to 2501.63 ± 1021.21 s and increase the sleep rate from 58.33 to 83.33% in mice. Moreover, YPVEPF and YFYPEL could bind with the Ser-205 and Phe-77 residues of GABAAR via hydrogen bonds and lipid contacts. They were largely released after digestion with 11.19 ± 0.15 and 1.78 ± 0.01 g/kg, respectively.
Collapse
Affiliation(s)
- Jingjing Qian
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Guowan Su
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Mingtao Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Donghui Luo
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521000, China
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521000, China
- Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| |
Collapse
|
10
|
Nonclinical pharmacology of daridorexant: a new dual orexin receptor antagonist for the treatment of insomnia. Psychopharmacology (Berl) 2021; 238:2693-2708. [PMID: 34415378 PMCID: PMC8455402 DOI: 10.1007/s00213-021-05954-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/03/2021] [Indexed: 12/29/2022]
Abstract
Dual orexin receptor antagonists (DORAs) represent a novel type of sleep medication that provide an alternative to the traditionally used positive allosteric gamma-aminobutyric acid (GABA)-A receptor modulators. Daridorexant is a new DORA that exhibited in phase 3 trials in insomnia not only a beneficial effect on sleep variables, measured objectively and assessed subjectively, but also an improvement in daytime functioning. Daridorexant was discovered through a tailored research program aimed at identifying an optimized sleep-promoting molecule with pharmacokinetic properties appropriate for covering the whole night while avoiding next-morning residual activity at efficacious doses. By specific binding to both orexin receptors, daridorexant inhibits the actions of the wake-promoting orexin (also called hypocretin) neuropeptides. This mechanism avoids a more widespread inhibition of neuronal pathways and associated side effects that are intrinsic to positive allosteric GABA-A receptor modulators. Here, we review the general pharmacology of daridorexant, based on nonclinical pharmacology studies of daridorexant, unpublished or already described, or based on work with other DORAs. Some unique features of daridorexant will be highlighted, such as the promotion of natural and surmountable sleep, the preservation of memory and cognition, the absence of tolerance development or risk of physical dependence, and how it can benefit daytime functioning.
Collapse
|
11
|
Abstract
The scope of this article is to review the effects on sleep of prescription drugs that are commonly prescribed for chronic insomnia in adults. The following groups are discussed: benzodiazepines and its receptor agonists, the dual orexin receptor antagonist suvorexant, melatonin and its receptor agonists, sedating antidepressants, and antipsychotics. Together with the neurobiologic and pharmacologic properties of these drugs, clinical effects are described, including subjective and objective effects on sleep duration, continuity, and architecture. Medical prescription information is given when available. Recently published American and European guidelines for the treatment of insomnia serve as reference frame.
Collapse
Affiliation(s)
- Sylvie Dujardin
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands
| | - Angelique Pijpers
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands
| | - Dirk Pevernagie
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| |
Collapse
|
12
|
Satyanarayanan SK, Chien YC, Chang JPC, Huang SY, Guu TW, Su H, Su KP. Melatonergic agonist regulates circadian clock genes and peripheral inflammatory and neuroplasticity markers in patients with depression and anxiety. Brain Behav Immun 2020; 85:142-151. [PMID: 30851380 DOI: 10.1016/j.bbi.2019.03.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE Circadian dysfunction is a core manifestation and a risk factor for psychiatric disorders. Ramelteon (RMT), a melatonin receptor agonist, has been shown to induce sleep phase shifts and has been used to normalize sleep onset time. RMT has been used in sleep disorders, depression and anxiety. In this study, we aimed to investigate the effects of RMT in regulating gene expression profiles of the circadian clock and peripheral markers of inflammation and neuroplasticity. METHODS Sixteen patients with a diagnosis of primary insomnia comorbid with depression and anxiety and ten healthy controls were recruited in an 8-week open-label trial. The patients with primary insomnia received RMT 8 mg/day. The morning expression profiles of 15 core clock genes from peripheral blood mononuclear cells (PBMCs), urine and plasma levels of melatonin and its metabolite levels, and plasma inflammatory markers and neurotrophin levels were evaluated at baseline, 4th and 8th week of RMT treatment. RESULTS RMT treatment was associated with significant clinical improvement in depression scores at 8th week (Hamilton depression rating scale scores (Mean ± SEM) from 21.5 ± 2.44 to 14.31 ± 2.25, p ≤ 0.05). The overall poor sleep quality (Pittsburgh sleep quality index) of the patient group significantly improved (p ≤ 0.05) following RMT treatment. The mRNA level analysis showed a significant association between RMT treatment and alterations of the nine core circadian genes (CLOCK, PER1, PER2, CRY1, CRY2, NR1D1, NR1D2, DEC1 and TIMELESS) in the patient group when compared with the control group (p ≤ 0.05). Compared with the controls, the patient group had a decrease in neurotrophins (brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor and beta-nerve growth factor; p ≤ 0.05) but an increase in pro-inflammatory cytokine levels (interleukin-6, interleukin-1b, tumour necrosis factor-alpha and interferon gamma; p ≤ 0.05); RMT treatment normalized the levels of neurotrophins and cytokine levels. CONCLUSION RMT treatment is able to restore phase-shifted melatonin markers, normalized the altered expression of the circadian genes, the levels of inflammatory cytokines and neurotrophins in patients with insomnia comorbid anxiety and depression.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yu-Chuan Chien
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Division of Psychiatry, Departments of Internal Medicine, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Jane Pei-Chen Chang
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; College of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan
| | - Ta-Wei Guu
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Division of Psychiatry, Departments of Internal Medicine, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Kuan-Pin Su
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
13
|
Belelli D, Hogenkamp D, Gee KW, Lambert JJ. Realising the therapeutic potential of neuroactive steroid modulators of the GABA A receptor. Neurobiol Stress 2019; 12:100207. [PMID: 32435660 PMCID: PMC7231973 DOI: 10.1016/j.ynstr.2019.100207] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/19/2019] [Indexed: 01/27/2023] Open
Abstract
In the 1980s particular endogenous metabolites of progesterone and of deoxycorticosterone were revealed to be potent, efficacious, positive allosteric modulators (PAMs) of the GABAA receptor (GABAAR). These reports were followed by the discovery that such steroids may be synthesised not only in peripheral endocrine glands, but locally in the central nervous system (CNS), to potentially act as paracrine, or autocrine "neurosteroid" messengers, thereby fine tuning neuronal inhibition. These discoveries triggered enthusiasm to elucidate the physiological role of such neurosteroids and explore whether their levels may be perturbed in particular psychiatric and neurological disorders. In preclinical studies the GABAAR-active steroids were shown to exhibit anxiolytic, anticonvulsant, analgesic and sedative properties and at relatively high doses to induce a state of general anaesthesia. Collectively, these findings encouraged efforts to investigate the therapeutic potential of neurosteroids and related synthetic analogues. However, following over 30 years of investigation, realising their possible medical potential has proved challenging. The recent FDA approval for the natural neurosteroid allopregnanolone (brexanolone) to treat postpartum depression (PPD) should trigger renewed enthusiasm for neurosteroid research. Here we focus on the influence of neuroactive steroids on GABA-ergic signalling and on the challenges faced in developing such steroids as anaesthetics, sedatives, analgesics, anticonvulsants, antidepressants and as treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Delia Belelli
- Systems Medicine, Neuroscience, Mail Box 6, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, United Kingdom
| | - Derk Hogenkamp
- Department of Pharmacology, 110C Med Surge1, Mail Code 4625, University of California, Irvine, School of Medicine, Irvine, CA, 92697, USA
| | - Kelvin W Gee
- Department of Pharmacology, 110C Med Surge1, Mail Code 4625, University of California, Irvine, School of Medicine, Irvine, CA, 92697, USA
| | - Jeremy J Lambert
- Systems Medicine, Neuroscience, Mail Box 6, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, United Kingdom
| |
Collapse
|
14
|
Dornbierer DA, Baur DM, Stucky B, Quednow BB, Kraemer T, Seifritz E, Bosch OG, Landolt HP. Neurophysiological signature of gamma-hydroxybutyrate augmented sleep in male healthy volunteers may reflect biomimetic sleep enhancement: a randomized controlled trial. Neuropsychopharmacology 2019; 44:1985-1993. [PMID: 30959514 PMCID: PMC6785068 DOI: 10.1038/s41386-019-0382-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/25/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Gamma-hydroxybutyrate (GHB) is an endogenous GHB/GABAB receptor agonist, which has demonstrated potency in consolidating sleep and reducing excessive daytime sleepiness in narcolepsy. Little is known whether GHB's efficacy reflects the promotion of physiological sleep mechanisms and no study has investigated its sleep consolidating effects under low sleep pressure. GHB (50 mg/kg p.o.) and placebo were administered in 20 young male volunteers at 2:30 a.m., the time when GHB is typically given in narcolepsy, in a randomized, double-blinded, crossover manner. Drug effects on sleep architecture and electroencephalographic (EEG) sleep spectra were analyzed. In addition, current source density (CSD) analysis was employed to identify the effects of GHB on the brain electrical sources of neuronal oscillations. Moreover, lagged-phase synchronization (LPS) analysis was applied to quantify the functional connectivity among sleep-relevant brain regions. GHB prolonged slow-wave sleep (stage N3) at the cost of rapid eye movement (REM) sleep. Furthermore, it enhanced delta-theta (0.5-8 Hz) activity in NREM and REM sleep, while reducing activity in the spindle frequency range (13-15 Hz) in sleep stage N2. The increase in delta power predominated in medial prefrontal cortex, parahippocampal and fusiform gyri, and posterior cingulate cortex. Theta power was particularly increased in the prefrontal cortex and both temporal poles. Moreover, the brain areas that showed increased theta power after GHB also exhibited increased lagged-phase synchronization among each other. Our study in healthy men revealed distinct similarities between GHB-augmented sleep and physiologically augmented sleep as seen in recovery sleep after prolonged wakefulness. The promotion of the sleep neurophysiological mechanisms by GHB may thus provide a rationale for GHB-induced sleep and waking quality in neuropsychiatric disorders beyond narcolepsy.
Collapse
Affiliation(s)
- Dario A Dornbierer
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zürich, Zürich, Switzerland.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Lenggstrasse 31, Zürich, CH-8032, Switzerland.
| | - Diego M Baur
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Sleep & Health Zürich, University Center of Competence, University of Zürich, Zürich, Switzerland
| | - Benjamin Stucky
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Sleep & Health Zürich, University Center of Competence, University of Zürich, Zürich, Switzerland
| | - Boris B Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Lenggstrasse 31, Zürich, CH-8032, Switzerland
| | - Thomas Kraemer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zürich, Zürich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Lenggstrasse 31, Zürich, CH-8032, Switzerland
- Sleep & Health Zürich, University Center of Competence, University of Zürich, Zürich, Switzerland
- HMZ Flagship SleepLoop of UZH and ETHZ, Zürich, Switzerland
| | - Oliver G Bosch
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Lenggstrasse 31, Zürich, CH-8032, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Sleep & Health Zürich, University Center of Competence, University of Zürich, Zürich, Switzerland
| |
Collapse
|
15
|
Abstract
Current GABAergic sleep-promoting medications were developed pragmatically, without making use of the immense diversity of GABAA receptors. Pharmacogenetic experiments are leading to an understanding of the circuit mechanisms in the hypothalamus by which zolpidem and similar compounds induce sleep at α2βγ2-type GABAA receptors. Drugs acting at more selective receptor types, for example, at receptors containing the α2 and/or α3 subunits expressed in hypothalamic and brain stem areas, could in principle be useful as hypnotics/anxiolytics. A highly promising sleep-promoting drug, gaboxadol, which activates αβδ-type receptors failed in clinical trials. Thus, for the time being, drugs such as zolpidem, which work as positive allosteric modulators at GABAA receptors, continue to be some of the most effective compounds to treat primary insomnia.
Collapse
Affiliation(s)
- W Wisden
- Department Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| | - X Yu
- Department Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - N P Franks
- Department Life Sciences, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
16
|
de Miguel E, Vekovischeva O, Elsilä LV, Panhelainen A, Kankuri E, Aitta-Aho T, Korpi ER. Conditioned Aversion and Neuroplasticity Induced by a Superagonist of Extrasynaptic GABA A Receptors: Correlation With Activation of the Oval BNST Neurons and CRF Mechanisms. Front Mol Neurosci 2019; 12:130. [PMID: 31178693 PMCID: PMC6543524 DOI: 10.3389/fnmol.2019.00130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/03/2019] [Indexed: 11/13/2022] Open
Abstract
THIP (gaboxadol), a superagonist of the δ subunit-containing extrasynaptic GABAA receptors, produces persistent neuroplasticity in dopamine (DA) neurons of the ventral tegmental area (VTA), similarly to rewarding drugs of abuse. However, unlike them THIP lacks abuse potential and induces conditioned place aversion in mice. The mechanism underlying the aversive effects of THIP remains elusive. Here, we show that mild aversive effects of THIP were detected 2 h after administration likely reflecting an anxiety-like state with increased corticosterone release and with central recruitment of corticotropin-releasing factor corticotropin-releasing factor receptor 1 (CRF1) receptors. A detailed immunohistochemical c-Fos expression mapping for THIP-activated brain areas revealed a correlation between the activation of CRF-expressing neurons in the oval nucleus of the bed nuclei of stria terminalis and THIP-induced aversive effects. In addition, the neuroplasticity of mesolimbic DA system (24 h after administration) and conditioned place aversion by THIP after four daily acute sessions were dependent on extrasynaptic GABAA receptors (abolished in δ-GABAA receptor knockout mice) and activation of the CRF1 receptors (abolished in wildtype mice by a CRF1 receptor antagonist). A selective THIP-induced activation of CRF-expressing neurons in the oval part of the bed nucleus of stria terminalis may constitute a novel mechanism for inducing plasticity in a population of VTA DA neurons and aversive behavioral states.
Collapse
Affiliation(s)
- Elena de Miguel
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olga Vekovischeva
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri V Elsilä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
17
|
Korkutata M, Saitoh T, Cherasse Y, Ioka S, Duo F, Qin R, Murakoshi N, Fujii S, Zhou X, Sugiyama F, Chen JF, Kumagai H, Nagase H, Lazarus M. Enhancing endogenous adenosine A2A receptor signaling induces slow-wave sleep without affecting body temperature and cardiovascular function. Neuropharmacology 2019; 144:122-132. [DOI: 10.1016/j.neuropharm.2018.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/10/2018] [Accepted: 10/14/2018] [Indexed: 01/20/2023]
|
18
|
|
19
|
Shelton KT, Qu J, Bilotta F, Brown EN, Cudemus G, D’Alessandro DA, Deng H, DiBiasio A, Gitlin JA, Hahm EY, Hobbs LE, Houle TT, Ibala R, Loggia M, Pavone KJ, Shaefi S, Tolis G, Westover MB, Akeju O. Minimizing ICU Neurological Dysfunction with Dexmedetomidine-induced Sleep (MINDDS): protocol for a randomised, double-blind, parallel-arm, placebo-controlled trial. BMJ Open 2018; 8:e020316. [PMID: 29678977 PMCID: PMC5914725 DOI: 10.1136/bmjopen-2017-020316] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Delirium, which is prevalent in postcardiac surgical patients, is an acute brain dysfunction characterised by disturbances in attention, awareness and cognition not explained by a pre-existing neurocognitive disorder. The pathophysiology of delirium remains poorly understood. However, basic science and clinical studies suggest that sleep disturbance may be a modifiable risk factor for the development of delirium. Dexmedetomidine is a α-2A adrenergic receptor agonist medication that patterns the activity of various arousal nuclei similar to sleep. A single night-time loading dose of dexmedetomidine promotes non-rapid eye movement sleep stages N2 and N3 sleep. This trial hypothesises dexmedetomidine-induced sleep as pre-emptive therapy for postoperative delirium. METHODS AND ANALYSIS The MINDDS (Minimizing ICU Neurological Dysfunction with Dexmedetomidine-induced Sleep) trial is a 370-patient block-randomised, placebo-controlled, double-blinded, single-site, parallel-arm superiority trial. Patients over 60 years old, undergoing cardiac surgery with planned cardiopulmonary bypass, will be randomised to receive a sleep-inducing dose of dexmedetomidine or placebo. The primary outcome is the incidence of delirium on postoperative day 1, assessed with the Confusion Assessment Method by staff blinded to the treatment assignment. To ensure that the study is appropriately powered for the primary outcome measure, patients will be recruited and randomised into the study until 370 patients receive the study intervention on postoperative day 0. Secondary outcomes will be evaluated by in-person assessments and medical record review for in-hospital end points, and by telephone interview for 30-day, 90-day and 180-day end points. All trial outcomes will be evaluated using an intention-to-treat analysis plan. Hypothesis testing will be performed using a two-sided significance level (type I error) of α=0.05. Sensitivity analyses using the actual treatment received will be performed and compared with the intention-to-treat analysis results. Additional sensitivity analyses will assess the potential impact of missing data due to loss of follow-up. ETHICS AND DISSEMINATION The Partners Human Research Committee approved the MINDDS trial. Recruitment began in March 2017. Dissemination plans include presentations at scientific conferences, scientific publications and popular media. TRIAL REGISTRATION NUMBER NCT02856594.
Collapse
Affiliation(s)
- Kenneth T Shelton
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jason Qu
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Federico Bilotta
- Department of Anaesthesia and Critical Care Medicine, Sapienza University of Rome, Rome, Italy
| | - Emery N Brown
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Gaston Cudemus
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A D’Alessandro
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hao Deng
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alan DiBiasio
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jacob A Gitlin
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eunice Y Hahm
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lauren E Hobbs
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Timothy T Houle
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reine Ibala
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marco Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kara J Pavone
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shahzad Shaefi
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - George Tolis
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - M. Brandon Westover
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Oluwaseun Akeju
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Hong KB, Park Y, Suh HJ. Two combined amino acids promote sleep activity in caffeine-induced sleepless model systems. Nutr Res Pract 2018; 12:208-214. [PMID: 29854326 PMCID: PMC5974066 DOI: 10.4162/nrp.2018.12.3.208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/08/2018] [Accepted: 03/13/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND/OBJECTIVES The aim of this study was to evaluate the biological and sleep-promoting effects of combined γ-aminobutyric acid (GABA) and 5-hydroxytryptophan (5-HTP) using caffeine-induced sleepless fruit flies, ICR mice, and Sprague-Dawley rats. MATERIALS/METHODS Video-tracking analysis was applied to investigate behavioral changes of Drosophila melanogaster. Pentobarbital-induced sleep test and electroencephalogram (EEG) patterns were used for analysis of sleep latency, duration, and quantity and quality of sleep in vertebrate models. RESULTS Administration of combined GABA/5-HTP could significantly reverse the caffeine induced total distance of flies (P < 0.001). Also, individually administered and combined GABA/5-HTP significantly increased the total sleeping time in the caffeine-induced sleepless ICR mice (P < 0.001). In the caffeine-induced sleepless SD-rats, combined GABA/5-HTP showed significant differences in sleep quality between individual amino acid administrations (P < 0.05). CONCLUSIONS Taken together, we identified inhibitory effects of combined GABA/5-HTP in locomotor activity, sleep quantity and quality in caffeine-induced sleepless models, indicating that combined GABA/5-HTP may be effective in patients with insomnia by providing sufficient sleep.
Collapse
Affiliation(s)
- Ki-Bae Hong
- Department of Biological Sciences and Environmental Sciences Program, Southern Illinois University-Edwardsville, Edwardsville, IL 62026, United States
| | - Yooheon Park
- Dongguk University Research Institute of Biotechnology and Medical Converged Science, Dongguk University, Gyeonggi 10325, Korea
| | - Hyung Joo Suh
- Department of Public Health Sciences, Korea University, 145 Anam-ro, Sungbuk-gu, Seoul 02841, Korea
| |
Collapse
|
21
|
Atkin T, Comai S, Gobbi G. Drugs for Insomnia beyond Benzodiazepines: Pharmacology, Clinical Applications, and Discovery. Pharmacol Rev 2018; 70:197-245. [PMID: 29487083 DOI: 10.1124/pr.117.014381] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although the GABAergic benzodiazepines (BZDs) and Z-drugs (zolpidem, zopiclone, and zaleplon) are FDA-approved for insomnia disorders with a strong evidence base, they have many side effects, including cognitive impairment, tolerance, rebound insomnia upon discontinuation, car accidents/falls, abuse, and dependence liability. Consequently, the clinical use of off-label drugs and novel drugs that do not target the GABAergic system is increasing. The purpose of this review is to analyze the neurobiological and clinical evidence of pharmacological treatments of insomnia, excluding the BZDs and Z-drugs. We analyzed the melatonergic agonist drugs, agomelatine, prolonged-release melatonin, ramelteon, and tasimelteon; the dual orexin receptor antagonist suvorexant; the modulators of the α2δ subunit of voltage-sensitive calcium channels, gabapentin and pregabalin; the H1 antagonist, low-dose doxepin; and the histamine and serotonin receptor antagonists, amitriptyline, mirtazapine, trazodone, olanzapine, and quetiapine. The pharmacology and mechanism of action of these treatments and the evidence-base for the use of these drugs in clinical practice is outlined along with novel pipelines. There is evidence to recommend suvorexant and low-dose doxepin for sleep maintenance insomnia; there is also sufficient evidence to recommend ramelteon for sleep onset insomnia. Although there is limited evidence for the use of the quetiapine, trazodone, mirtazapine, amitriptyline, pregabalin, gabapentin, agomelatine, and olanzapine as treatments for insomnia disorder, these drugs may improve sleep while successfully treating comorbid disorders, with a different side effect profile than the BZDs and Z-drugs. The unique mechanism of action of each drug allows for a more personalized and targeted medical management of insomnia.
Collapse
Affiliation(s)
- Tobias Atkin
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada (T.A., S.C., G.G.); and Division of Neuroscience, San Raffaele Scientific Institute and Vita-Salute University, Milan, Italy (S.C.)
| |
Collapse
|
22
|
Akeju O, Hobbs LE, Gao L, Burns SM, Pavone KJ, Plummer GS, Walsh EC, Houle TT, Kim SE, Bianchi MT, Ellenbogen JM, Brown EN. Dexmedetomidine promotes biomimetic non-rapid eye movement stage 3 sleep in humans: A pilot study. Clin Neurophysiol 2017; 129:69-78. [PMID: 29154132 DOI: 10.1016/j.clinph.2017.10.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/26/2017] [Accepted: 10/11/2017] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Sleep, which comprises of rapid eye movement (REM) and non-REM stages 1-3 (N1-N3), is a natural occurring state of decreased arousal that is crucial for normal cardiovascular, immune and cognitive function. The principal sedative drugs produce electroencephalogram beta oscillations, which have been associated with neurocognitive dysfunction. Pharmacological induction of altered arousal states that neurophysiologically approximate natural sleep, termed biomimetic sleep, may eliminate drug-induced neurocognitive dysfunction. METHODS We performed a prospective, single-site, three-arm, randomized-controlled, crossover polysomnography pilot study (n = 10) comparing natural, intravenous dexmedetomidine- (1-μg/kg over 10 min [n = 7] or 0.5-μg/kg over 10 min [n = 3]), and zolpidem-induced sleep in healthy volunteers. Sleep quality and psychomotor performance were assessed with polysomnography and the psychomotor vigilance test, respectively. Sleep quality questionnaires were also administered. RESULTS We found that dexmedetomidine promoted N3 sleep in a dose dependent manner, and did not impair performance on the psychomotor vigilance test. In contrast, zolpidem extended release was associated with decreased theta (∼5-8 Hz; N2 and N3) and increased beta oscillations (∼13-25 Hz; N2 and REM). Zolpidem extended release was also associated with increased lapses on the psychomotor vigilance test. No serious adverse events occurred. CONCLUSIONS Pharmacological induction of biomimetic N3 sleep with psychomotor sparing benefits is feasible. SIGNIFICANCE These results suggest that α2a adrenergic agonists may be developed as a new class of sleep enhancing medications with neurocognitive sparing benefits.
Collapse
Affiliation(s)
- Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lauren E Hobbs
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara M Burns
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kara J Pavone
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - George S Plummer
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisa C Walsh
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tim T Houle
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Seong-Eun Kim
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Electronics and Control Engineering, Hanbat National University, Daejon, Republic of Korea
| | - Matt T Bianchi
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Emery N Brown
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
23
|
Bottom-Up versus Top-Down Induction of Sleep by Zolpidem Acting on Histaminergic and Neocortex Neurons. J Neurosci 2017; 36:11171-11184. [PMID: 27807161 DOI: 10.1523/jneurosci.3714-15.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 08/17/2016] [Indexed: 12/18/2022] Open
Abstract
Zolpidem, a GABAA receptor-positive modulator, is the gold-standard drug for treating insomnia. Zolpidem prolongs IPSCs to decrease sleep latency and increase sleep time, effects that depend on α2 and/or α3 subunit-containing receptors. Compared with natural NREM sleep, zolpidem also decreases the EEG power, an effect that depends on α1 subunit-containing receptors, and which may make zolpidem-induced sleep less optimal. In this paper, we investigate whether zolpidem needs to potentiate only particular GABAergic pathways to induce sleep without reducing EEG power. Mice with a knock-in F77I mutation in the GABAA receptor γ2 subunit gene are zolpidem-insensitive. Using these mice, GABAA receptors in the frontal motor neocortex and hypothalamic (tuberomammillary nucleus) histaminergic-neurons of γ2I77 mice were made selectively sensitive to zolpidem by genetically swapping the γ2I77 subunits with γ2F77 subunits. When histamine neurons were made selectively zolpidem-sensitive, systemic administration of zolpidem shortened sleep latency and increased sleep time. But in contrast to the effect of zolpidem on wild-type mice, the power in the EEG spectra of NREM sleep was not decreased, suggesting that these EEG power-reducing effects of zolpidem do not depend on reduced histamine release. Selective potentiation of GABAA receptors in the frontal cortex by systemic zolpidem administration also reduced sleep latency, but less so than for histamine neurons. These results could help with the design of new sedatives that induce a more natural sleep. SIGNIFICANCE STATEMENT Many people who find it hard to get to sleep take sedatives. Zolpidem (Ambien) is the most widely prescribed "sleeping pill." It makes the inhibitory neurotransmitter GABA work better at its receptors throughout the brain. The sleep induced by zolpidem does not resemble natural sleep because it produces a lower power in the brain waves that occur while we are sleeping. We show using mouse genetics that zolpidem only needs to work on specific parts and cell types of the brain, including histamine neurons in the hypothalamus, to induce sleep but without reducing the power of the sleep. This knowledge could help in the design of sleeping pills that induce a more natural sleep.
Collapse
|
24
|
Drakakis G, Wafford KA, Brewerton SC, Bodkin MJ, Evans DA, Bender A. Polypharmacological in Silico Bioactivity Profiling and Experimental Validation Uncovers Sedative-Hypnotic Effects of Approved and Experimental Drugs in Rat. ACS Chem Biol 2017; 12:1593-1602. [PMID: 28414209 DOI: 10.1021/acschembio.7b00209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this work, we describe the computational ("in silico") mode-of-action analysis of CNS-active drugs, which is taking both multiple simultaneous hypotheses as well as sets of protein targets for each mode-of-action into account, and which was followed by successful prospective in vitro and in vivo validation. Using sleep-related phenotypic readouts describing both efficacy and side effects for 491 compounds tested in rat, we defined an "optimal" (desirable) sleeping pattern. Compounds were subjected to in silico target prediction (which was experimentally confirmed for 21 out of 28 cases), followed by the utilization of decision trees for deriving polypharmacological bioactivity profiles. We demonstrated that predicted bioactivities improved classification performance compared to using only structural information. Moreover, DrugBank molecules were processed via the same pipeline, and compounds in many cases not annotated as sedative-hypnotic (alcaftadine, benzatropine, palonosetron, ecopipam, cyproheptadine, sertindole, and clopenthixol) were prospectively validated in vivo. Alcaftadine, ecopipam cyproheptadine, and clopenthixol were found to promote sleep as predicted, benzatropine showed only a small increase in NREM sleep, whereas sertindole promoted wakefulness. To our knowledge, the sedative-hypnotic effects of alcaftadine and ecopipam have not been previously discussed in the literature. The method described extends previous single-target, single-mode-of-action models and is applicable across disease areas.
Collapse
Affiliation(s)
- Georgios Drakakis
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Keith A. Wafford
- Eli Lilly U.K., Erl Wood Manor, Windlesham, Surrey GU206PH, United Kingdom
| | | | - Michael J. Bodkin
- Eli Lilly U.K., Erl Wood Manor, Windlesham, Surrey GU206PH, United Kingdom
| | - David A. Evans
- Eli Lilly U.K., Erl Wood Manor, Windlesham, Surrey GU206PH, United Kingdom
| | - Andreas Bender
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
25
|
De Crescenzo F, Foti F, Ciabattini M, Del Giovane C, Watanabe N, Sañé Schepisi M, Quested DJ, Cipriani A, Barbui C, Amato L. Comparative efficacy and acceptability of pharmacological treatments for insomnia in adults: a systematic review and network meta-analysis. Hippokratia 2016. [DOI: 10.1002/14651858.cd012364] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Franco De Crescenzo
- Catholic University of the Sacred Heart; Institute of Psychiatry and Psychology; L.go A. Gemelli 8 Rome Italy 00168
| | - Francesca Foti
- "Sapienza" University of Rome; Department of Psychology; Via dei Marsi 78 Rome Italy 00185
- IRCCS Santa Lucia Foundation; Via del Fosso di Fiorano 64 Rome Italy 00143
| | | | - Cinzia Del Giovane
- University of Modena and Reggio Emilia; Italian Cochrane Centre, Department of Diagnostic, Clinical and Public Health Medicine; Modena Italy
| | - Norio Watanabe
- Kyoto University Graduate School of Medicine / School of Public Health; Department of Health Promotion and Human Behavior; Yoshida Konoe-cho, Sakyo-ku Kyoto Kyoto Japan 606-8501
| | - Monica Sañé Schepisi
- National Institute for Infectious Diseases, IRCCS L. Spallanzani; Clinical Epidemiology; Rome, 00149 Italy
| | - Digby J Quested
- Oxford Health NHS Trust; Department of Psychiatry and Mental Health; Oxford UK
| | - Andrea Cipriani
- University of Oxford; Department of Psychiatry; Warneford Hospital Oxford UK OX3 7JX
| | - Corrado Barbui
- University of Verona; Neuroscience, Biomedicine and Movement Sciences, Section of Psychiatry; Verona Italy
| | - Laura Amato
- Lazio Regional Health Service; Department of Epidemiology; Via Cristoforo Colombo, 112 Rome Italy 00154
| |
Collapse
|
26
|
Heidmann B, Gatfield J, Roch C, Treiber A, Tortoioli S, Brotschi C, Williams JT, Bolli MH, Abele S, Sifferlen T, Jenck F, Boss C. Discovery of Highly Potent Dual Orexin Receptor Antagonists via a Scaffold-Hopping Approach. ChemMedChem 2016; 11:2132-2146. [DOI: 10.1002/cmdc.201600175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/18/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Bibia Heidmann
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - John Gatfield
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Catherine Roch
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Alexander Treiber
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Simone Tortoioli
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Christine Brotschi
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Jodi T. Williams
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Martin H. Bolli
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Stefan Abele
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Thierry Sifferlen
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - François Jenck
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| | - Christoph Boss
- Actelion Pharmaceuticals Ltd.; Drug Discovery and Preclinical Research & Development; Gewerbestrasse 16 4123 Allschwil Switzerland
| |
Collapse
|
27
|
Talele TT. The "Cyclopropyl Fragment" is a Versatile Player that Frequently Appears in Preclinical/Clinical Drug Molecules. J Med Chem 2016; 59:8712-8756. [PMID: 27299736 DOI: 10.1021/acs.jmedchem.6b00472] [Citation(s) in RCA: 624] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recently, there has been an increasing use of the cyclopropyl ring in drug development to transition drug candidates from the preclinical to clinical stage. Important features of the cyclopropane ring are, the (1) coplanarity of the three carbon atoms, (2) relatively shorter (1.51 Å) C-C bonds, (3) enhanced π-character of C-C bonds, and (4) C-H bonds are shorter and stronger than those in alkanes. The present review will focus on the contributions that a cyclopropyl ring makes to the properties of drugs containing it. Consequently, the cyclopropyl ring addresses multiple roadblocks that can occur during drug discovery such as (a) enhancing potency, (b) reducing off-target effects,
Collapse
Affiliation(s)
- Tanaji T Talele
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University , 8000 Utopia Parkway, Queens, New York 11439, United States
| |
Collapse
|
28
|
GABA Receptors on Orexin and Melanin-Concentrating Hormone Neurons Are Differentially Homeostatically Regulated Following Sleep Deprivation. eNeuro 2016; 3:eN-NWR-0077-16. [PMID: 27294196 PMCID: PMC4899679 DOI: 10.1523/eneuro.0077-16.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/21/2022] Open
Abstract
Though overlapping in distribution through the hypothalamus, orexin (Orx) and melanin-concentrating hormone (MCH) neurons play opposite roles in the regulation of sleep-wake states. Orx neurons discharge during waking, whereas MCH neurons discharge during sleep. In the present study, we examined in mice whether GABAA and GABAB receptors (Rs) are present on Orx and MCH neurons and might undergo differential changes as a function of their different activities following sleep deprivation (SD) and sleep recovery (SR). Applying quantitative stereological image analysis to dual-immunofluorescent stained sections, we determined that the proportion of Orx neurons positively immunostained for GABAARs was significantly higher following SD (∼48%) compared with sleep control (SC; ∼24%) and SR (∼27%), and that the luminance of the GABAARs was significantly greater. In contrast, the average proportion of the MCH neurons immunostained for GABAARs was insignificantly lower following SD (∼43%) compared with SC (∼54%) and SR (56%), and the luminance of the GABAARs was significantly less. Although, GABABRs were observed in all Orx and MCH neurons (100%), the luminance of these receptors was differentially altered following SD. The intensity of GABABRs in the Orx neurons was significantly greater after SD than after SC and SR, whereas that in the MCH neurons was significantly less. The present results indicate that GABA receptors undergo dynamic and differential changes in the wake-active Orx neurons and the sleep-active MCH neurons as a function of and homeostatic adjustment to their preceding activity and sleep-wake state.
Collapse
|
29
|
Offord J, Isom LL. Drugging the undruggable: gabapentin, pregabalin and the calcium channel α2δ subunit. Crit Rev Biochem Mol Biol 2016; 51:246-56. [DOI: 10.3109/10409238.2016.1173010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Martin BS, Martinez-Botella G, Loya CM, Salituro FG, Robichaud AJ, Huntsman MM, Ackley MA, Doherty JJ, Corbin JG. Rescue of deficient amygdala tonic γ-aminobutyric acidergic currents in the Fmr-/y mouse model of fragile X syndrome by a novel γ-aminobutyric acid type A receptor-positive allosteric modulator. J Neurosci Res 2015; 94:568-78. [PMID: 26308557 DOI: 10.1002/jnr.23632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 11/07/2022]
Abstract
Alterations in the ratio of excitatory to inhibitory transmission are emerging as a common component of many nervous system disorders, including autism spectrum disorders (ASDs). Tonic γ-aminobutyric acidergic (GABAergic) transmission provided by peri- and extrasynaptic GABA type A (GABAA ) receptors powerfully controls neuronal excitability and plasticity and, therefore, provides a rational therapeutic target for normalizing hyperexcitable networks across a variety of disorders, including ASDs. Our previous studies revealed tonic GABAergic deficits in principal excitatory neurons in the basolateral amygdala (BLA) in the Fmr1(-/y) knockout (KO) mouse model fragile X syndrome. To correct amygdala deficits in tonic GABAergic neurotransmission in Fmr1(-/y) KO mice, we developed a novel positive allosteric modulator of GABAA receptors, SGE-872, based on endogenously active neurosteroids. This study shows that SGE-872 is nearly as potent and twice as efficacious for positively modulating GABAA receptors as its parent molecule, allopregnanolone. Furthermore, at submicromolar concentrations (≤1 μM), SGE-872 is selective for tonic, extrasynaptic α4β3δ-containing GABAA receptors over typical synaptic α1β2γ2 receptors. We further find that SGE-872 strikingly rescues the tonic GABAergic transmission deficit in principal excitatory neurons in the Fmr1(-/y) KO BLA, a structure heavily implicated in the neuropathology of ASDs. Therefore, the potent and selective action of SGE-872 on tonic GABAA receptors containing α4 subunits may represent a novel and highly useful therapeutic avenue for ASDs and related disorders involving hyperexcitability of neuronal networks.
Collapse
Affiliation(s)
- Brandon S Martin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| | | | - Carlos M Loya
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | | | - Molly M Huntsman
- Pharmaceutical Sciences and Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Mike A Ackley
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| |
Collapse
|
31
|
Effects of blue mussel (ME) water extracts on pentobarbital-induced sleep and the sleep architecture in mice. Food Sci Biotechnol 2015. [DOI: 10.1007/s10068-015-0039-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
32
|
Doxepin and diphenhydramine increased non-rapid eye movement sleep through blockade of histamine H1 receptors. Pharmacol Biochem Behav 2015; 129:56-64. [DOI: 10.1016/j.pbb.2014.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/04/2014] [Accepted: 12/08/2014] [Indexed: 11/23/2022]
|
33
|
Pharmacological evaluation of sedative and hypnotic effects of schizandrin through the modification of pentobarbital-induced sleep behaviors in mice. Eur J Pharmacol 2014; 744:157-63. [DOI: 10.1016/j.ejphar.2014.09.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 08/24/2014] [Accepted: 09/04/2014] [Indexed: 01/22/2023]
|
34
|
May AC, Fleischer W, Kletke O, Haas HL, Sergeeva OA. Benzodiazepine-site pharmacology on GABAA receptors in histaminergic neurons. Br J Pharmacol 2014; 170:222-32. [PMID: 23799902 DOI: 10.1111/bph.12280] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The histaminergic tuberomamillary nucleus (TMN) of the posterior hypothalamus controls the cognitive aspects of vigilance which is reduced by common sedatives and anxiolytics. The receptors targeted by these drugs in histaminergic neurons are unknown. TMN neurons express nine different subunits of the GABAA receptor (GABAA R) with three α- (α1, α2 and α5) and two γ- (γ1, γ 2) subunits, which confer different pharmacologies of the benzodiazepine-binding site. EXPERIMENTAL APPROACH We investigated the actions of zolpidem, midazolam, diazepam, chlordiazepoxide, flumazenil (Ro15-1788) and methyl-6,7-dimethoxy-4-ethyl-β-carboline-3-carboxylate (DMCM) in TMN neurons using mouse genetics, electrophysiological and molecular biological methods. KEY RESULTS We find the sensitivity of GABAA R to zolpidem, midazolam and DMCM significantly reduced in TMN neurons from γ2F77I mice, but modulatory activities of diazepam, chlordiazepoxide and flumazenil not affected. Potencies and efficacies of these compounds are in line with the dominance of α2- and α1-subunit containing receptors associated with γ2- or γ1-subunits. Functional expression of the γ1-subunit is supported by siRNA-based knock-down experiments in γ2F77I mice. CONCLUSIONS AND IMPLICATIONS GABAA R of TMN neurons respond to a variety of common sedatives with a high affinity binding site (γ2F77I) involved. The γ1-subunit likely contributes to the action of common sedatives in TMN neurons. This study is relevant for understanding the role of neuronal histamine and benzodiazepines in disorders of sleep and metabolism.
Collapse
Affiliation(s)
- A C May
- Department of Neurophysiology, Medical Faculty, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
35
|
Shah VK, Choi JJ, Han JY, Lee MK, Hong JT, Oh KW. Pachymic Acid Enhances Pentobarbital-Induced Sleeping Behaviors via GABAA-ergic Systems in Mice. Biomol Ther (Seoul) 2014; 22:314-20. [PMID: 25143810 PMCID: PMC4131518 DOI: 10.4062/biomolther.2014.045] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 05/22/2014] [Accepted: 06/13/2014] [Indexed: 11/21/2022] Open
Abstract
This study was investigated to know whether pachymic acid (PA), one of the predominant triterpenoids in Poria cocos (Hoelen) has the sedative-hypnotic effects, and underlying mechanisms are mediated via γ-aminobutyric acid (GABA)-ergic systems. Oral administration of PA markedly suppressed locomotion activity in mice. This compound also prolonged sleeping time, and reduced sleep latency showing synergic effects with muscimol (0.2 mg/kg) in shortening sleep onset and enhancing sleep time induced by pentobarbital, both at the hypnotic (40 mg/kg) and sub-hypnotic (28 mg/kg) doses. Additionally, PA elevated intracellular chloride levels in hypothalamic primary cultured neuronal cells of rats. Moreover, Western blotting quantitative results showed that PA increased the amount of protein level expression of GAD65/67 over a broader range of doses. PA increased α- and β-subunits protein levels, but decreased γ-subunit protein levels in GABAA receptors. The present experiment provides evidence for the hypnotic effects as PA enhanced pentobarbital-induced sleeping behaviors via GABAA-ergic mechanisms in rodents. Taken together, it is proposed that PA may be useful for the treatment of sleep disturbed subjects with insomnia.
Collapse
Affiliation(s)
- Vikash Kumar Shah
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Jae Joon Choi
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Jin-Yi Han
- Institute of Veterinary Medicine, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Mi Kyeong Lee
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Ki-Wan Oh
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Republic of Korea
| |
Collapse
|
36
|
Zhang C, Mao X, Zhao X, Liu Z, Liu B, Li H, Bi K, Jia Y. Gomisin N isolated from Schisandra chinensis augments pentobarbital-induced sleep behaviors through the modification of the serotonergic and GABAergic system. Fitoterapia 2014; 96:123-30. [DOI: 10.1016/j.fitote.2014.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022]
|
37
|
Xu XH, Qiu MH, Dong H, Qu WM, Urade Y, Huang ZL. GABA transporter-1 inhibitor NO-711 alters the EEG power spectra and enhances non-rapid eye movement sleep during the active phase in mice. Eur Neuropsychopharmacol 2014; 24:585-94. [PMID: 24080505 DOI: 10.1016/j.euroneuro.2013.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 09/02/2013] [Accepted: 09/05/2013] [Indexed: 11/25/2022]
Abstract
GABA transporter subtype 1 (GAT1) constructs high affinity reuptake sites for GABA in the CNS and regulates GABAergic transmission. Compounds that inhibit GAT1 are targets often used for the treatment of epilepsy; however sedation has been reported as a side effect of these agents, indicating potential sedative and/or hypnotic uses for these compounds. In the current study, we observed the sleep behaviors of mice treated with NO-711, a selective GAT1 inhibitor, in order to elucidate the role of GAT1 in sleep-wake regulation during the active phase. The data revealed that NO-711 at a high dose of 10 mg/kg caused a marked enhancement of EEG activity in the frequency ranges of 3-25 Hz during wakefulness as well as rapid eye movement (REM) sleep. During the non-REM (NREM) sleep, NO-711 (10 mg/kg) elevated EEG activity in the frequency ranges of 1.5-6.75 Hz. Similar changes were found in mice treated with a low dose of 3 mg/kg. NO-711 administered i.p. at a dose of 1, 3 or 10 mg/kg significantly shortened the sleep latency of NREM sleep, increased the amount of NREM sleep and the number of NREM sleep episodes. NO-711 did not affect the sleep latency and the amount of REM sleep. NO-711 dose-dependently increased c-Fos expression in sleep-promoting nucleus of the ventrolateral preoptic area and median preoptic area. However, c-Fos expression was decreased in the wake-promoting nuclei, tuberomammillary nucleus and lateral hypothalamus. These results indicate that NO-711 can increase NREM sleep in mice.
Collapse
Affiliation(s)
- Xin-Hong Xu
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mei-Hong Qiu
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Hui Dong
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, China; Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Yoshihiro Urade
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Suita, Osaka, Japan
| | - Zhi-Li Huang
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China; Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Neuroscience-driven discovery and development of sleep therapeutics. Pharmacol Ther 2014; 141:300-34. [DOI: 10.1016/j.pharmthera.2013.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 10/25/2013] [Indexed: 01/18/2023]
|
39
|
Li M, Kang R, Jia S, Shi J, Liu G, Zhang J. Sedative and hypnotic activity of N(6)-(3-methoxyl-4-hydroxybenzyl) adenine riboside (B2), an adenosine analog. Pharmacol Biochem Behav 2013; 117:151-6. [PMID: 24361595 DOI: 10.1016/j.pbb.2013.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 12/10/2013] [Accepted: 12/13/2013] [Indexed: 11/26/2022]
Abstract
N(6)-(3-methoxyl-4-hydroxybenzyl) adenine riboside (B2) is an N(6)-substitued adenosine analog. Previous studies have shown that B2 binds to the adenosine A1 and A2A receptors with moderate affinity and produces protective effects on serum deprivation-induced cell damage. However, central nervous system effects of B2 have not been studied. We aimed to investigate the sedative and hypnotic effects and the mechanism of action of B2 in mice. Our behavioral studies showed that oral administration of B2 decreased spontaneous locomotor activity and potentiated the hypnotic effect of pentobarbital in mice. Sleep architecture analyses revealed that B2 decreased wakefulness and increased non-rapid eye movement (NREM) sleep in both normal mice and mice with caffeine-induced insomnia. Using immunohistochemistry, we showed that B2 increased c-Fos expression, a cellular marker for neuronal activity, in the ventrolateral preoptic (VLPO) area, a sleep center in the anterior hypothalamus. Altogether, these results indicate that oral administration of B2 produces sedative and hypnotic effects. Furthermore, the activation of VLPO neurons may be involved in the central depressant effects of B2.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Department of Clinical Pharmacology, Beijing Hospital of the Ministry of Health, Beijing 100730, PR China
| | - Ruixia Kang
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Shaobo Jia
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Jiangong Shi
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - GengTao Liu
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - JianJun Zhang
- State Key Laboratory Of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
40
|
Tou WI, Chen CYC. May disordered protein cause serious drug side effect? Drug Discov Today 2013; 19:367-72. [PMID: 24184432 DOI: 10.1016/j.drudis.2013.10.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 09/23/2013] [Accepted: 10/25/2013] [Indexed: 01/16/2023]
Abstract
Insomnia is a self-reported disease where patients lose their ability to initiate and maintain sleep, leading to daytime performance impairment. Several drug targets to ameliorate insomnia symptoms have been discovered; however, these drug targets lead to serious side effects. Thus, we characterize the structural properties of these sleep-related receptors and the clock complex and discuss a possible drug design that will reduce side effects. Computational prediction shows that disordered property is shared. Over 30% of the structure of CLOCK, PER1/2/3, BMAL-1, muscarinic acetylcholine receptor-M1, melatonin receptor and casein kinase I are structurally disordered (the remaining proteins represent <30%). Investigations support the principle that the failures of insomnia drugs might be closely related to the protein architecture.
Collapse
Affiliation(s)
- Weng Ieong Tou
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Calvin Yu-Chian Chen
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan; Department of Biotechnology, Asia University, Taichung, 41354, Taiwan; China Medical University Beigang Hospital, Yunlin, 65152, Taiwan; Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
41
|
Krystal AD, Richelson E, Roth T. Review of the histamine system and the clinical effects of H1 antagonists: Basis for a new model for understanding the effects of insomnia medications. Sleep Med Rev 2013; 17:263-72. [DOI: 10.1016/j.smrv.2012.08.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 11/27/2022]
|
42
|
Tolerability, pharmacokinetics, and pharmacodynamics of single-dose almorexant, an orexin receptor antagonist, in healthy elderly subjects. J Clin Psychopharmacol 2013; 33:363-70. [PMID: 23609389 DOI: 10.1097/jcp.0b013e31828f5a7a] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sleep disorders are common in the elderly population. Orexin receptor antagonism has been proposed as a new sleep-enabling approach to treat insomnia. The tolerability, pharmacokinetics, and pharmacodynamics of ascending single doses of almorexant, a dual orexin receptor antagonist, were investigated in healthy elderly male and female subjects. In this double-blind, placebo- and active-controlled study, each dose (100, 200, and 400 mg) was investigated in a separate group of 12 subjects (almorexant, placebo, and zolpidem 10 mg in an 8:2:2 ratio). Morning doses of almorexant were well tolerated. As expected for sleep-enabling compounds, somnolence and fatigue were frequently reported. Other adverse events included headache and nausea. Muscular weakness was reported at a higher incidence only with the highest almorexant dose. The pharmacokinetic profile of almorexant was characterized by a median time to the maximum concentration of 1.5 hours, quick disposition with a distribution half-life of 1.6 hours, and rapidly decreasing concentrations to approximately 20% of the maximum concentration over 8 hours, with a terminal half-life of 32 hours. Objective pharmacodynamic measures showed decreases in saccadic peak velocity and adaptive tracking performance and increases in body sway with the 400-mg dose of almorexant. Subjective assessments revealed a dose-dependent decrease in alertness. Almorexant had no effects on mood, calmness, subjective internal and external perception, and feeling high. These findings provide a solid basis to study the effects of almorexant in elderly patients with insomnia.
Collapse
|
43
|
Abstract
Sleep disturbance during menopause is a common and important complaint faced by many women. There are many factors that may play a role in this problem, including vasomotor symptoms and changing hormone levels, circadian rhythm abnormalities, exacerbation of primary insomnia, mood disorders, coexistent medical conditions as well as lifestyle factors. Sleep can be measured both objectively and subjectively; however, correlation between the two measures is not high. Most of the menopause-related sleep disturbances have been reported as qualitative in nature; however, there have also been studies showing changes in objective measures. This discrepancy has implications with regard to evaluation of research in sleep and menopause, as well as application in the clinical setting. Investigations of inadequate sleep and sleep problems during the menopausal period and obtaining a thorough understanding of the factors contributing to these problems are essential in formulating treatment strategies. Such strategies can vary from hormonal treatment and medications to lifestyle and behavioural modification.
Collapse
Affiliation(s)
- D Ameratunga
- Department of Obstetrics & Gynecology, The Royal Women's Hospital, Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
44
|
Camargos EF, Louzada FM, Nóbrega OT. Wrist actigraphy for measuring sleep in intervention studies with Alzheimer's disease patients: application, usefulness, and challenges. Sleep Med Rev 2013; 17:475-88. [PMID: 23669093 DOI: 10.1016/j.smrv.2013.01.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 01/26/2013] [Accepted: 01/28/2013] [Indexed: 10/26/2022]
Abstract
Sleep disorders are common in patients with Alzheimer's disease (AD). An important aspect of intervention studies in patients with sleep disorders is the choice of assessment strategy. This paper presents a literature review concerning assessment strategies for measuring sleep in intervention studies with AD patients, with a focus on actigraphy. Thirty-seven articles were selected for this review, having analysis of sleep/nocturnal rhythm disturbances by actigraphy as the primary or secondary outcome. The advantages and limitations of actigraphy were discussed vis-à-vis polysomnography and subjective interventions. The following methodological aspects were addressed: impact of experimental design and patient setting, inclusion and exclusion criteria, placement of the actigraphy device, adherence to the regimen, duration of recordings and the choice of sleep parameters. Our analyses suggest that the methods used in intervention studies encompassing sleep disorders and dementia could be improved by increasing accuracy of diagnosis, categorization of sleep disturbances, adherence to actigraphy, and by clearly defining the variables and endpoints in each study. Also, controlling variables that could interfere with sleep and describing the data processing and analysis might improve interpretation of results.
Collapse
Affiliation(s)
- Einstein F Camargos
- Department of Medical Clinical, Geriatric Medical Centre, HUB, Brasilia University Hospital, SGAN 605 Av. L2 Norte, Asa Norte, Brasilia, DF, CEP 70840-901, Brazil.
| | | | | |
Collapse
|
45
|
Dingemanse J, Hoever P, Hoch M, Treiber A, Wagner-Redeker W, Miraval T, Hopfgartner G, Shakeri-Nejad K. Elucidation of the metabolic pathways and the resulting multiple metabolites of almorexant, a dual orexin receptor antagonist, in humans. Drug Metab Dispos 2013; 41:1046-59. [PMID: 23431113 DOI: 10.1124/dmd.112.050120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Almorexant [(2R)-2-{(1S)-6, 7-dimethoxy-1-[2-(4-trifluoromethyl-phenyl)-ethyl]-3,4-dihydro-1H-isoquinolin-2-yl}-N-methyl-2-phenyl-acetamide], a tetrahydroisoquinoline derivative, is a dual orexin receptor antagonist with sleep-promoting properties in both animals and humans. This study investigated the disposition, metabolism, and elimination of almorexant in humans. After oral administration of a 200-mg dose of ¹⁴C-almorexant, almorexant was rapidly absorbed (Tmax = 0.8 hour), and the apparent terminal half-life (t(1/2)) was 17.8 hours. The radioactive dose was almost completely recovered with 78.0% of the administered radioactive dose found in feces and 13.5% in urine. Unchanged almorexant was not found in urine and represented 10% of the administered dose in feces. In total, 47 metabolites were identified of which 21 were shown to be present in plasma. There are four primary metabolites, the isomeric phenols M3 and M8, formed by demethylation, the aromatic isoquinolinium ion M5, formed by dehydrogenation, and M6, formed by oxidative dealkylation with loss of the phenylglycine moiety. Most of the subsequent products are formed by permutations of these primary metabolic reactions followed by conjugation of the intermediate phenols with glucuronic or sulfonic acid. The percentage of dose excreted in urine or feces for any of the metabolites did not exceed 10% of the administered radioactive dose, nor did any of the metabolites represent more than 10% of the total drug-related exposure. In conclusion, after rapid absorption, almorexant is extensively metabolized, and excretion of metabolites in feces is the predominant route of elimination in humans.
Collapse
Affiliation(s)
- Jasper Dingemanse
- Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
GABAergic inhibition of histaminergic neurons regulates active waking but not the sleep-wake switch or propofol-induced loss of consciousness. J Neurosci 2012; 32:13062-75. [PMID: 22993424 DOI: 10.1523/jneurosci.2931-12.2012] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The activity of histaminergic neurons in the tuberomammillary nucleus (TMN) of the hypothalamus correlates with an animal's behavioral state and maintains arousal. We examined how GABAergic inputs onto histaminergic neurons regulate this behavior. A prominent hypothesis, the "flip-flop" model, predicts that increased and sustained GABAergic drive onto these cells promotes sleep. Similarly, because of the histaminergic neurons' key hub-like place in the arousal circuitry, it has also been suggested that anesthetics such as propofol induce loss of consciousness by acting primarily at histaminergic neurons. We tested both these hypotheses in mice by genetically removing ionotropic GABA(A) or metabotropic GABA(B) receptors from histidine decarboxylase-expressing neurons. At the cellular level, histaminergic neurons deficient in synaptic GABA(A) receptors were significantly more excitable and were insensitive to the anesthetic propofol. At the behavioral level, EEG profiles were recorded in nontethered mice over 24 h. Surprisingly, GABAergic transmission onto histaminergic neurons had no effect in regulating the natural sleep-wake cycle and, in the case of GABA(A) receptors, for propofol-induced loss of righting reflex. The latter finding makes it unlikely that the histaminergic TMN has a central role in anesthesia. GABA(B) receptors on histaminergic neurons were dispensable for all behaviors examined. Synaptic inhibition of histaminergic cells by GABA(A) receptors, however, was essential for habituation to a novel environment.
Collapse
|
47
|
Flavonoid Myricetin Modulates GABA(A) Receptor Activity through Activation of Ca(2+) Channels and CaMK-II Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:758097. [PMID: 23258999 PMCID: PMC3520426 DOI: 10.1155/2012/758097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/31/2012] [Accepted: 08/10/2012] [Indexed: 11/17/2022]
Abstract
The flavonoid myricetin is found in several sedative herbs, for example, the St. John's Wort, but its influence on sedation and its possible mechanism of action are unknown. Using patch-clamp technique on a brain slice preparation, the present study found that myricetin promoted GABAergic activity in the neurons of hypothalamic paraventricular nucleus (PVN) by increasing the decay time and frequency of the inhibitory currents mediated by GABAA receptor. This effect of myricetin was not blocked by the GABAA receptor benzodiazepine- (BZ-) binding site antagonist flumazenil, but by KN-62, a specific inhibitor of the Ca2+/calmodulin-stimulated protein kinase II (CaMK-II). Patch clamp and live Ca2+ imaging studies found that myricetin could increase Ca2+ current and intracellular Ca2+ concentration, respectively, via T- and L-type Ca2+ channels in rat PVN neurons and hypothalamic primary culture neurons. Immunofluorescence staining showed increased phosphorylation of CaMK-II after myricetin incubation in primary culture of rat hypothalamic neurons, and the myricetin-induced CaMK-II phosphorylation was further confirmed by Western blotting in PC-12 cells. The present results suggest that myricetin enhances GABAA receptor activity via calcium channel/CaMK-II dependent mechanism, which is distinctively different from that of most existing BZ-binding site agonists of GABAA receptor.
Collapse
|
48
|
Hoever P, de Haas SL, Dorffner G, Chiossi E, van Gerven JM, Dingemanse J. Orexin receptor antagonism: an ascending multiple-dose study with almorexant. J Psychopharmacol 2012; 26:1071-80. [PMID: 22695489 DOI: 10.1177/0269881112448946] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The objectives of this study were to investigate the multiple-dose tolerability, safety, pharmacokinetics, and pharmacodynamics of the dual orexin receptor antagonist almorexant. Healthy subjects received daily doses of almorexant (100, 200, 400 or 1000 mg) or placebo in the morning for four days followed by two days with evening administration (Days 5-6). Each dose level was investigated in a new group of 10 subjects (eight active, two placebo, 1:1 sex). Dose-dependent increases in frequency and intensity were observed for somnolence and other adverse events. Pharmacokinetics at steady state showed rapid absorption, low concentrations eight hours post-dose, and minimal accumulation. Following evening, administration absorption was delayed and C(max) decreased. Almorexant at 400 and 1000 mg administered in the morning reduced vigilance, alertness, visuomotor coordination, and motor coordination assessed in a psychometric test battery. Polysomnography recordings following evening administration showed a trend towards shorter latency to sleep stages 3 and 4, and shorter latency to, and longer time in, rapid-eye-movement sleep at higher doses when compared to placebo. Whether these findings in healthy subjects translate into relevant sleep-enabling effects in insomnia patients needs to be investigated in future studies.
Collapse
Affiliation(s)
- Petra Hoever
- Department ofClinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, Allschwil, Switzerland
| | | | | | | | | | | |
Collapse
|
49
|
Brickley SG, Mody I. Extrasynaptic GABA(A) receptors: their function in the CNS and implications for disease. Neuron 2012; 73:23-34. [PMID: 22243744 DOI: 10.1016/j.neuron.2011.12.012] [Citation(s) in RCA: 519] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2011] [Indexed: 12/30/2022]
Abstract
Over the past two decades, research has identified extrasynaptic GABA(A) receptor populations that enable neurons to sense the low ambient GABA concentrations present in the extracellular space in order to generate a form of tonic inhibition not previously considered in studies of neuronal excitability. The importance of this tonic inhibition in regulating states of consciousness is highlighted by the fact that extrasynaptic GABA(A) receptors (GABA(A)Rs) are believed to be key targets for anesthetics, sleep-promoting drugs, neurosteroids, and alcohol. The neurosteroid sensitivity of these extrasynaptic GABA(A)Rs may explain their importance in stress-, ovarian cycle-, and pregnancy-related mood disorders. Moreover, disruptions in network dynamics associated with schizophrenia, epilepsy, and Parkinson's disease may well involve alterations in the tonic GABA(A)R-mediated conductance. Extrasynaptic GABA(A)Rs may therefore present a therapeutic target for treatment of these diseases, with the potential to enhance cognition and aid poststroke functional recovery.
Collapse
Affiliation(s)
- Stephen G Brickley
- Division of Cell & Molecular Biology, South Kensington Campus, Imperial College, London SW7 2AZ, UK.
| | | |
Collapse
|
50
|
Anaclet C, Zhang M, Zhao C, Buda C, Seugnet L, Lin JS. Effects of GF-015535-00, a novel α1 GABA A receptor ligand, on the sleep-wake cycle in mice, with reference to zolpidem. Sleep 2012; 35:103-11. [PMID: 22215924 DOI: 10.5665/sleep.1596] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Novel, safe, and efficient hypnotic compounds capable of enhancing physiological sleep are still in great demand in the therapy of insomnia. This study compares the sleep-wake effects of a new α1 GABA(A) receptor subunit ligand, GF-015535-00, with those of zolpidem, the widely utilized hypnotic compound. METHODS Nine C57Bl6/J male mice were chronically implanted with electrodes for EEG and sleep-wake monitoring. Each mouse received 3 doses of GF-015535-00 and zolpidem. Time spent in sleep-wake states and cortical EEG power spectra were analyzed. RESULTS Both zolpidem and GF-015535-00 prominently enhanced slow wave sleep and paradoxical sleep in the mouse. However, as compared with zolpidem, GF-015535-00 showed several important differences: (1) a comparable sleep-enhancing effect was obtained with a 10 fold smaller dose; (2) the induced sleep was less fragmented; (3) the risk of subsequent wake rebound was less prominent; and (4) the cortical EEG power ratio between slow wave sleep and wake was similar to that of natural sleep and thus compatible with physiological sleep. CONCLUSION The characteristics of the sleep-wake effects of GF-015535-00 in mice could be potentially beneficial for its use as a therapeutic compound in the treatment of insomnia. Further investigations are required to assess whether the same characteristics are conserved in other animal models and humans.
Collapse
Affiliation(s)
- Christelle Anaclet
- Integrative Physiology of Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, 8 Avenue Rockefeller, Lyon Cedex 08, France
| | | | | | | | | | | |
Collapse
|