1
|
Huang L, Zhu W, Li N, Zhang B, Dai W, Li S, Xu H. Functions and mechanisms of adenosine and its receptors in sleep regulation. Sleep Med 2024; 115:210-217. [PMID: 38373361 DOI: 10.1016/j.sleep.2024.02.012] [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: 11/06/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/21/2024]
Abstract
Sleep is a natural and recurring state of life. Long-term insomnia can lead to physical and mental fatigue, inattention, memory loss, anxiety, depression and other symptoms, imposing immense public health and economic burden worldwide. The sleep and awakening regulation system is composed of many nerve nuclei and neurotransmitters in the brain, and it forms a neural network that interacts and restricts each other to regulate the occurrence and maintenance of sleep-wake. Adenosine (AD) is a neurotransmitter in the central nervous system and a driver of sleep. Meanwhile, the functions and mechanisms underlying sleep-promoting effects of adenosine and its receptors are still not entirely clear. However, in recent years, the increasing evidence indicated that adenosine can promote sleep through inhibiting arousal system and activating sleep-promoting system. At the same time, astrocyte-derived adenosine in modulating sleep homeostasis and sleep loss-induced related cognitive and memory deficits plays an important role. This review, therefore, summarizes the current research on the functions and possible mechanisms of adenosine and its receptors in the regulation of sleep and homeostatic control of sleep. Understanding these aspects will provide us better ideas on clinical problems such as insomnia, hypersomnia and other sleep disorders.
Collapse
Affiliation(s)
- Lishan Huang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenwen Zhu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Nanxi Li
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Bin Zhang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Wenbin Dai
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China.
| | - Houping Xu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
2
|
Wolfson AR, Wong D, Abrams EM, Waserman S, Sussman GL. Diphenhydramine: Time to Move on? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:3124-3130. [PMID: 35999169 DOI: 10.1016/j.jaip.2022.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022]
Abstract
Diphenhydramine is one of the most widely available, longest-used antihistamine medications but has many side effects including sedation and risk of toxicity in overdose including cardiac toxicity. It is frequently inappropriately used when newer, more favorable antihistamine medications are available. Second-generation antihistamines are also widely available and affordable, with many of the same desired effects as diphenhydramine and fewer, if any, of the undesirable side effects. Because of the negative side effects and wide availability of alternative antihistamine medications, it is definitively time to move on from diphenhydramine.
Collapse
Affiliation(s)
- Anna R Wolfson
- Harvard Medical School, Boston, Mass; Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass.
| | - Dennis Wong
- Division of Clinical Immunology and Allergy, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Elissa M Abrams
- University of Manitoba, Department of Pediatrics, Section of Allergy and Clinical Immunology, Winnipeg, MB, Canada; University of British Columbia, Department of Pediatrics, Division of Allergy and Immunology, Vancouver, BC, Canada
| | - Susan Waserman
- Division of Clinical Immunology and Allergy, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Gordon L Sussman
- Division of Clinical Immunology and Allergy, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Patel R, Agrawal S, Jain NS. Stimulation of dorsal hippocampal histaminergic transmission mitigates the expression of ethanol withdrawal-induced despair in mice. Alcohol 2021; 96:1-14. [PMID: 34228989 DOI: 10.1016/j.alcohol.2021.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/12/2021] [Accepted: 06/25/2021] [Indexed: 10/20/2022]
Abstract
Garnered literature points toward the role of the dorsal hippocampus (CA1) in ethanol withdrawal-induced responses, wherein a strong presence of the histaminergic system is also reported. Therefore, the present study investigated the effect of an enhanced CA1 histaminergic transmission on the expression of chronic ethanol withdrawal-induced despair in mice on the tail suspension test (TST). The results revealed that mice who were on an ethanol-fed diet (5.96%, v/v) for 8 days exhibited maximum immobility time on the TST, and decreased locomotion at 24 h post-ethanol withdrawal (10th day), indicating ethanol withdrawal-induced despair. Enhancement of CA1 histaminergic activity achieved by the treatment of intra-CA1 microinjection of histaminergic agents such as histamine (0.1, 10 μg/mouse, bilateral), the histamine precursor l-histidine (1, 10 μg/mouse, bilateral), the histamine neuronal releaser/H3 receptor antagonist thioperamide (2, 10 μg/mouse, bilateral), the histamine H1 receptor agonist FMPH (2, 6.5 μg/mouse, bilateral), or the H2 receptor agonist amthamine (0.1, 0.5 μg/mouse, bilateral) to ethanol-withdrawn mice, 10 min before the 24-h post-ethanol withdrawal time point, significantly alleviated the expression of ethanol withdrawal-induced despair in mice on the TST. On the other hand, only the pre-treatment of the histamine H1 receptor agonist FMPH (2, 6.5 μg/mouse, intra-CA1 bilateral) reversed the reduction in locomotor activity induced in ethanol-withdrawn mice, whereas other employed histaminergic agents were devoid of any effect on this behavior. Therefore, our findings indicate that an enhanced CA1 histaminergic transmission, probably via stimulation of CA1 postsynaptic histamine H1 or H2 receptor, could preclude the behavioral despair, while H1 stimulation affects motor deficit expressed after ethanol withdrawal.
Collapse
|
4
|
Bellot-Saez A, Stevenson R, Kékesi O, Samokhina E, Ben-Abu Y, Morley JW, Buskila Y. Neuromodulation of Astrocytic K + Clearance. Int J Mol Sci 2021; 22:ijms22052520. [PMID: 33802343 PMCID: PMC7959145 DOI: 10.3390/ijms22052520] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022] Open
Abstract
Potassium homeostasis is fundamental for brain function. Therefore, effective removal of excessive K+ from the synaptic cleft during neuronal activity is paramount. Astrocytes play a key role in K+ clearance from the extracellular milieu using various mechanisms, including uptake via Kir channels and the Na+-K+ ATPase, and spatial buffering through the astrocytic gap-junction coupled network. Recently we showed that alterations in the concentrations of extracellular potassium ([K+]o) or impairments of the astrocytic clearance mechanism affect the resonance and oscillatory behavior of both the individual and networks of neurons. These results indicate that astrocytes have the potential to modulate neuronal network activity, however, the cellular effectors that may affect the astrocytic K+ clearance process are still unknown. In this study, we have investigated the impact of neuromodulators, which are known to mediate changes in network oscillatory behavior, on the astrocytic clearance process. Our results suggest that while some neuromodulators (5-HT; NA) might affect astrocytic spatial buffering via gap-junctions, others (DA; Histamine) primarily affect the uptake mechanism via Kir channels. These results suggest that neuromodulators can affect network oscillatory activity through parallel activation of both neurons and astrocytes, establishing a synergistic mechanism to maximize the synchronous network activity.
Collapse
Affiliation(s)
- Alba Bellot-Saez
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Rebecca Stevenson
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Orsolya Kékesi
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Evgeniia Samokhina
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yuval Ben-Abu
- Projects and Physics Section, Sapir Academic College, D.N. Hof Ashkelon, Sderot 79165, Israel;
| | - John W. Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (A.B.-S.); (R.S.); (O.K.); (E.S.); (J.W.M.)
- International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW 2751, Australia
- Correspondence: ; Tel.: +61-246203853
| |
Collapse
|
5
|
Mori T, Uzawa N, Masukawa D, Hirayama S, Iwase Y, Hokazono M, Udagawa Y, Suzuki T. Enhancement of the rewarding effects of 3,4-methylenedioxymethamphetamine in orexin knockout mice. Behav Brain Res 2020; 396:112802. [PMID: 32653557 DOI: 10.1016/j.bbr.2020.112802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/07/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022]
Abstract
Orexinergic neurons, which are closely associated with narcolepsy, regulate arousal and reward circuits through the activation of monoaminergic neurons. Psychostimulants as well as 5-HT-related compounds have potential in the treatment of human narcolepsy. Previous studies have demonstrated that orexin receptor antagonists as well as orexin deficiencies affect the pharmacological effects of psychostimulants. However, little information is available on the consequences of psychostimulant use under orexin deficiency. Therefore, the present study was designed to investigate the abuse liability of psychostimulants in orexin knockout (KO) mice. In the present study, conditioned place preferences induced by methamphetamine and methylphenidate were not altered in orexin KO mice. Interestingly, we found that MDMA induced a conditioned place preference in orexin KO mice, but not in wild type (WT) mice. In addition, MDMA produced methylphenidate/methamphetamine-like discriminative stimulus effects in orexin KO mice, but not WT mice. Increases in 5-HT and dopamine release in the nucleus accumbens induced by MDMA were not altered by knockout of orexin; the steady-state level of G protein activation was higher in the limbic forebrain of orexin KO mice. In substitution tests using a drug discrimination procedure, substitution of 5-HT1A receptor agonist for the discriminative stimulus effects of methylphenidate was enhanced in orexin KO mice. These findings indicate that the orexinergic system is involved the rewarding effects of psychostimulants. However, there is a risk of establishing rewarding effects of psychostimulants even under orexin deficiency. On the other hand, deficiencies in orexin may enhance the abuse liability of MDMA by changing a postsynaptic signal transduction accompanied by changes in discriminative stimulus effects themselves.
Collapse
Affiliation(s)
- Tomohisa Mori
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan; Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan.
| | - Naoki Uzawa
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan; Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Daiki Masukawa
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Shigeto Hirayama
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Yoshiyuki Iwase
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan; Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Mayuna Hokazono
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Yuya Udagawa
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Tsutomu Suzuki
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan; Institute of Drug Addiction Research, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan.
| |
Collapse
|
6
|
Yamakawa T, Kurauchi Y, Hisatsune A, Seki T, Katsuki H. Endogenous Nitric Oxide Inhibits, Whereas Awakening Stimuli Increase, the Activity of a Subset of Orexin Neurons. Biol Pharm Bull 2019; 41:1859-1865. [PMID: 30504686 DOI: 10.1248/bpb.b18-00633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lateral hypothalamic area contains neurons expressing neuronal nitric oxide synthase (nNOS), in addition to orexin neurons. Here we examined whether the activity of orexin neurons was regulated by endogenous nitric oxide (NO) in male C57BL/6 mice. Caffeine (30 mg/kg, intraperitoneally (i.p.)) increased the number of orexin neurons positive for c-Fos, a marker of neuronal activity, and also increased the number of NOS/c-Fos-positive cells as identified by reduced nicotinamide adenine dinucleotide phosphate (NADPH) diaphorase histochemistry and c-Fos immunohistochemistry. Diphenhydramine hydrochloride (10 mg/kg. i.p.) decreased c-Fos-positive orexin neurons but had no significant effect on the number of c-Fos-positive NOS neurons. nNOS inhibitor 7-nitroindazole (25 mg/kg, i.p.) alone increased c-Fos-positive orexin neurons, and combined treatment with caffeine and 7-nitroindazole did not show additive effect in the number of c-Fos-positive orexin neurons. In contrast, 7-nitroindazole decreased c-Fos-positive NOS neurons and attenuated caffeine-induced increase in c-Fos-positive NOS neurons. Sleep deprivation increased c-Fos-positive cells in both orexin neurons and NOS neurons, and 7-nitroindazole did not show additive effect with sleep deprivation in the activation of orexin neurons. Together, these results suggest that endogenous NO negatively regulates the activity of a subset of orexin neurons, and this subset of orexin neurons overlaps with that activated by awakening stimuli.
Collapse
Affiliation(s)
- Takao Yamakawa
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Akinori Hisatsune
- Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program," Priority Organization for Innovation and Excellence, Kumamoto University
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
7
|
Noseda R, Borsook D, Burstein R. Neuropeptides and Neurotransmitters That Modulate Thalamo-Cortical Pathways Relevant to Migraine Headache. Headache 2018; 57 Suppl 2:97-111. [PMID: 28485844 DOI: 10.1111/head.13083] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/19/2022]
Abstract
Dynamic thalamic regulation of sensory signals allows the cortex to adjust better to rapidly changing behavioral, physiological, and environmental demands. To fulfill this role, thalamic neurons must themselves be subjected to constantly changing modulatory inputs that originate in multiple neurochemical pathways involved in autonomic, affective, and cognitive functions. This review defines a chemical framework for thinking about the complexity of factors that modulate the response properties of relay trigeminovascular thalamic neurons. Following the presentation of scientific evidence for monosynaptic connections between thalamic trigeminovascular neurons and axons containing glutamate, GABA, dopamine, noradrenaline, serotonin, histamine, orexin, and melanin-concentrating hormone, this review synthesizes a large body of data to propose that the transmission of headache-related nociceptive signals from the thalamus to the cortex is modulated by potentially opposing forces and that the so-called 'decision' of which system (neuropeptide/neurotransmitter) will dominate the firing of a trigeminovascular thalamic neuron at any given time is determined by the constantly changing physiological (sleep, wakefulness, food intake, body temperature, heart rate, blood pressure), behavioral (addiction, isolation), cognitive (attention, learning, memory use), and affective (stress, anxiety, depression, anger) adjustment needed to keep homeostasis.
Collapse
Affiliation(s)
- Rodrigo Noseda
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Borsook
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rami Burstein
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
|
9
|
Rapanelli M, Pittenger C. Histamine and histamine receptors in Tourette syndrome and other neuropsychiatric conditions. Neuropharmacology 2015; 106:85-90. [PMID: 26282120 DOI: 10.1016/j.neuropharm.2015.08.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/29/2015] [Accepted: 08/12/2015] [Indexed: 01/28/2023]
Abstract
The potential contributions of dysregulation of the brain's histaminergic modulatory system to neuropsychiatric disease, and the potential of histamine-targeting medications as therapeutic agents, are gradually coming into focus. The H3R receptor, which is expressed primarily in the central nervous system, is a promising pharmacotherapeutic target. Recent evidence for a contribution of histamine dysregulation to Tourette syndrome and tic disorders is particularly strong; although specific mutations in histamine-associated genes are rare, they have led to informative studies in animal models that may pave the way for therapeutic advances. A controlled study of an H3R antagonist in Tourette syndrome is ongoing. Preclinical studies of H3R antagonists in schizophrenia, attention deficit disorder, and narcolepsy have all shown promise. Recently reported controlled studies have been disappointing in schizophrenia and attention deficit disorder, but the H3R antagonist pitolisant shows promise in the treatment of narcolepsy and excessive daytime sleepiness and is currently under regulatory review for these conditions. This article is part of the Special Issue entitled 'Histamine Receptors'.
Collapse
Affiliation(s)
| | - Christopher Pittenger
- Department of Psychiatry, Yale University, New Haven, CT, USA; Department of Psychology, Yale University, New Haven, CT, USA; Department of Child Study Center, Yale University, New Haven, CT, USA; Department of Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA.
| |
Collapse
|
10
|
He C, Luo F, Chen X, Chen F, Li C, Ren S, Qiao Q, Zhang J, de Lecea L, Gao D, Hu Z. Superficial Layer-Specific Histaminergic Modulation of Medial Entorhinal Cortex Required for Spatial Learning. Cereb Cortex 2015; 26:1590-1608. [PMID: 25595181 DOI: 10.1093/cercor/bhu322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The medial entorhinal cortex (MEC) plays a crucial role in spatial learning and memory. Whereas the MEC receives a dense histaminergic innervation from the tuberomamillary nucleus of the hypothalamus, the functions of histamine in this brain region remain unclear. Here, we show that histamine acts via H1Rs to directly depolarize the principal neurons in the superficial, but not deep, layers of the MEC when recording at somata. Moreover, histamine decreases the spontaneous GABA, but not glutamate, release onto principal neurons in the superficial layers by acting at presynaptic H3Rs without effect on synaptic release in the deep layers. Histamine-induced depolarization is mediated via inhibition of Kir channels and requires the activation of protein kinase C, whereas the inhibition of spontaneous GABA release by histamine depends on voltage-gated Ca(2+) channels and extracellular Ca(2+). Furthermore, microinjection of the H1R or H3R, but not H2R, antagonist respectively into the superficial, but not deep, layers of MEC impairs rat spatial learning as assessed by water maze tasks but does not affect the motor function and exploratory activity in an open field. Together, our study indicates that histamine plays an essential role in spatial learning by selectively regulating neuronal excitability and synaptic transmission in the superficial layers of the MEC.
Collapse
Affiliation(s)
- Chao He
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Fenlan Luo
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Xingshu Chen
- Department of Histology and Embryology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Fang Chen
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Chao Li
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Shuancheng Ren
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Qicheng Qiao
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Jun Zhang
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, CA 94304, USA
| | - Dong Gao
- Department of Sleep and Psychology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Zhian Hu
- Department of Physiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| |
Collapse
|
11
|
Electric stimulation of the tuberomamillary nucleus affects epileptic activity and sleep-wake cycle in a genetic absence epilepsy model. Epilepsy Res 2014; 109:119-25. [PMID: 25524851 DOI: 10.1016/j.eplepsyres.2014.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/13/2014] [Accepted: 10/28/2014] [Indexed: 12/20/2022]
Abstract
Deep brain stimulation (DBS) is a promising approach for epilepsy treatment, but the optimal targets and parameters of stimulation are yet to be investigated. Tuberomamillary nucleus (TMN) is involved in EEG desynchronization-one of the proposed mechanisms for DBS action. We studied whether TMN stimulation could interfere with epileptic spike-wave discharges (SWDs) in WAG/Rij rats with inherited absence epilepsy and whether such stimulation would affect sleep-wake cycle. EEG and video registration were used to determine SWD occurrence and stages of sleep and wake during three-hours recording sessions. Stimulation (100Hz) was applied in two modes: closed-loop (with previously determined interruption threshold intensity) or open-loop mode (with 50% or 70% threshold intensity). Closed-loop stimulation successfully interrupted SWDs but elevated their number by 148 ± 54% compared to baseline. It was accompanied by increase in number of episodes but not total duration of both active and passive wakefulness. Open-loop stimulation with amplitude 50% threshold did not change measured parameters, though 70% threshold stimulation reduced SWDs number by 40 ± 9%, significantly raised the amount of active wakefulness and decreased the amount of both slow-wave and rapid eye movement sleep. These results suggest that the TMN is unfavorable as a target for DBS as its stimulation may cause alterations in sleep-wake cycle. A careful choosing of parameters and control of sleep-wake activity is necessary when applying DBS in epilepsy.
Collapse
|
12
|
Neurochemical pathways that converge on thalamic trigeminovascular neurons: potential substrate for modulation of migraine by sleep, food intake, stress and anxiety. PLoS One 2014; 9:e103929. [PMID: 25090640 PMCID: PMC4121288 DOI: 10.1371/journal.pone.0103929] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 07/08/2014] [Indexed: 12/24/2022] Open
Abstract
Dynamic thalamic regulation of sensory signals allows the cortex to adjust better to rapidly changing behavioral, physiological and environmental demands. To fulfill this role, thalamic neurons must themselves be subjected to constantly changing modulatory inputs that originate in multiple neurochemical pathways involved in autonomic, affective and cognitive functions. Our overall goal is to define an anatomical framework for conceptualizing how a ‘decision’ is made on whether a trigeminovascular thalamic neuron fires, for how long, and at what frequency. To begin answering this question, we determine which neuropeptides/neurotransmitters are in a position to modulate thalamic trigeminovascular neurons. Using a combination of in-vivo single-unit recording, juxtacellular labeling with tetramethylrhodamine dextran (TMR) and in-vitro immunohistochemistry, we found that thalamic trigeminovascular neurons were surrounded by high density of axons containing biomarkers of glutamate, GABA, dopamine and serotonin; moderate density of axons containing noradrenaline and histamine; low density of axons containing orexin and melanin concentrating hormone (MCH); but not axons containing CGRP, serotonin 1D receptor, oxytocin or vasopressin. In the context of migraine, the findings suggest that the transmission of headache-related nociceptive signals from the thalamus to the cortex may be modulated by opposing forces (i.e., facilitatory, inhibitory) that are governed by continuous adjustments needed to keep physiological, behavioral, cognitive and emotional homeostasis.
Collapse
|
13
|
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]
|
14
|
Rowe DL, Hermens DF. Attention-deficit/hyperactivity disorder: neurophysiology, information processing, arousal and drug development. Expert Rev Neurother 2014; 6:1721-34. [PMID: 17144785 DOI: 10.1586/14737175.6.11.1721] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this review, we draw on literature from both animal and human neurophysiological studies to consider the neurochemical mechanisms underlying attention-deficit/ hyperactivity disorder (ADHD). Psychophysiological and neuropsychological research is used to propose possible etiological endophenotypes of ADHD. These are conceptualized as patients with distinct cortical-arousal, information-processing or maturational abnormalities, or a combination thereof, and how the endophenotypes can be used to help drug development and optimize treatment and management. To illustrate, the paper focuses on neuro- and psychophysiological evidence that suggests cholinergic mechanisms may underlie specific information-processing abnormalities that occur in ADHD. The clinical implications for a cholinergic hypothesis of ADHD are considered, along with its possible implications for treatment and pharmacological development.
Collapse
Affiliation(s)
- Donald L Rowe
- The Brain Dynamics Centre and Department of Psychological Medicine, Westmead Hospital and University of Sydney, NSW, Australia.
| | | |
Collapse
|
15
|
Guryn R, Staszewski M, Walczyński K. Non-imidazole histamine H 3 ligands: part V. synthesis and preliminary pharmacological investigation of 1-[2-thiazol-4-yl- and 1-[2-thiazol-5-yl-(2-aminoethyl)]-4- n-propylpiperazine derivatives. Med Chem Res 2013; 22:3640-3652. [PMID: 23807824 PMCID: PMC3685697 DOI: 10.1007/s00044-012-0372-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 11/15/2012] [Indexed: 11/25/2022]
Abstract
Series of 1-[2-thiazol-4-yl-(2-aminoethyl)]- and 1-[2-thiazol-5-yl-(2-aminoethyl)]-4-n-propylpiperazine derivatives have been prepared and in vitro tested as H3-receptor antagonists (the electrically evoked contraction of the guinea-pig jejunum). It appeared that by comparison of homologous pairs, the 1-[2-thiazol-5-yl-(2-aminoethyl)]-4-n-propylpiperazines (3a,b and 4a-d) have much higher potency than their analogous 1-[2-thiazol-4-yl-(2-aminoethyl)]-4-n-propylpiperazines (2a-k). Based on the obtained results, we observed the 5-position of 2-methyl-2-R-aminoethyl substituents in the thiazole ring is favourable for histamine H3 receptor antagonist activity, whereas its presence in position 4 leads, almost in each case, to strong decrease of activity.
Collapse
Affiliation(s)
- Roman Guryn
- Department of Synthesis and Technology of Drugs, Medical University, Muszyńskiego Street 1, 90-145 Łódź, Poland
| | - Marek Staszewski
- Department of Synthesis and Technology of Drugs, Medical University, Muszyńskiego Street 1, 90-145 Łódź, Poland
| | - Krzysztof Walczyński
- Department of Synthesis and Technology of Drugs, Medical University, Muszyńskiego Street 1, 90-145 Łódź, Poland
| |
Collapse
|
16
|
|
17
|
Gondard E, Anaclet C, Akaoka H, Guo RX, Zhang M, Buda C, Franco P, Kotani H, Lin JS. Enhanced histaminergic neurotransmission and sleep-wake alterations, a study in histamine H3-receptor knock-out mice. Neuropsychopharmacology 2013; 38:1015-31. [PMID: 23303066 PMCID: PMC3629391 DOI: 10.1038/npp.2012.266] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Long-term abolition of a brain arousal system impairs wakefulness (W), but little is known about the consequences of long-term enhancement. The brain histaminergic arousal system is under the negative control of H3-autoreceptors whose deletion results in permanent enhancement of histamine (HA) turnover. In order to determine the consequences of enhancement of the histaminergic system, we compared the cortical EEG and sleep-wake states of H3-receptor knockout (H3R-/-) and wild-type mouse littermates. We found that H3R-/-mice had rich phenotypes. On the one hand, they showed clear signs of enhanced HA neurotransmission and vigilance, i.e., a higher EEG θ power during spontaneous W and a greater extent of W or sleep restriction during behavioral tasks, including environmental change, locomotion, and motivation tests. On the other hand, during the baseline dark period, they displayed deficient W and signs of sleep deterioration, such as pronounced sleep fragmentation and reduced cortical slow activity during slow wave sleep (SWS), most likely due to a desensitization of postsynaptic histaminergic receptors as a result of constant HA release. Ciproxifan (H3-receptor inverse agonist) enhanced W in wild-type mice, but not in H3R-/-mice, indicating a functional deletion of H3-receptors, whereas triprolidine (postsynaptic H1-receptor antagonist) or α-fluoromethylhistidine (HA-synthesis inhibitor) caused a greater SWS increase in H3R-/- than in wild-type mice, consistent with enhanced HA neurotransmission. These sleep-wake characteristics and the obesity phenotypes previously reported in this animal model suggest that chronic enhancement of histaminergic neurotransmission eventually compromises the arousal system, leading to sleep-wake, behavioral, and metabolic disorders similar to those caused by voluntary sleep restriction in humans.
Collapse
Affiliation(s)
- Elise Gondard
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Christelle Anaclet
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Hidéo Akaoka
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Rui-Xian Guo
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Mei Zhang
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Colette Buda
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France
| | | | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, Lyon, France,Integrative Physiology of the Brain Arousal Systems, Lyon Neuroscience Research Center, INSERM U1028-CNRS UMR 5292, Faculty of Medicine, Claude Bernard University, 8 av. Rockefeller, 69373, Lyon, Cedex 08 69373, France, Tel: (33) 478 777 116, Fax: (33) 478 777 150, E-mail:
| |
Collapse
|
18
|
Abstract
Switches between different behavioral states of the animal are associated with prominent changes in global brain activity, between sleep and wakefulness or from inattentive to vigilant states. What mechanisms control brain states, and what are the functions of the different states? Here we summarize current understanding of the key neural circuits involved in regulating brain states, with a particular emphasis on the subcortical neuromodulatory systems. At the functional level, arousal and attention can greatly enhance sensory processing, whereas sleep and quiet wakefulness may facilitate learning and memory. Several new techniques developed over the past decade promise great advances in our understanding of the neural control and function of different brain states.
Collapse
Affiliation(s)
- Seung-Hee Lee
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720
| |
Collapse
|
19
|
Zant JC, Rozov S, Wigren HK, Panula P, Porkka-Heiskanen T. Histamine release in the basal forebrain mediates cortical activation through cholinergic neurons. J Neurosci 2012; 32:13244-54. [PMID: 22993440 PMCID: PMC6621481 DOI: 10.1523/jneurosci.5933-11.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 08/02/2012] [Accepted: 08/02/2012] [Indexed: 01/13/2023] Open
Abstract
The basal forebrain (BF) is a key structure in regulating both cortical activity and sleep homeostasis. It receives input from all ascending arousal systems and is particularly highly innervated by histaminergic neurons. Previous studies clearly point to a role for histamine as a wake-promoting substance in the BF. We used in vivo microdialysis and pharmacological treatments in rats to study which electroencephalogram (EEG) spectral properties are associated with histamine-induced wakefulness and whether this wakefulness is followed by increased sleep and increased EEG delta power during sleep. We also investigated which BF neurons mediate histamine-induced cortical activation. Extracellular BF histamine levels rose immediately and remained constant throughout a 6 h period of sleep deprivation, returning to baseline levels immediately afterward. During the spontaneous sleep-wake cycle, we observed a strong correlation between wakefulness and extracellular histamine concentrations in the BF, which was unaffected by the time of day. The perfusion of histamine into the BF increased wakefulness and cortical activity without inducing recovery sleep. The perfusion of a histamine receptor 1 antagonist into the BF decreased both wakefulness and cortical activity. Lesioning the BF cholinergic neurons abolished these effects. Together, these results show that activation of the cholinergic BF by histamine is important in sustaining a high level of cortical activation, and that a lack of activation of the cholinergic BF by histamine may be important in initiating and maintaining nonrapid eye movement sleep. The level of histamine release is tightly connected to behavioral state, but conveys no information about sleep pressure.
Collapse
Affiliation(s)
- Janneke C. Zant
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, FIN-00014 Finland and
| | - Stanislav Rozov
- Neuroscience Center and Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, FIN-00014 Finland
| | - Henna-Kaisa Wigren
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, FIN-00014 Finland and
| | - Pertti Panula
- Neuroscience Center and Institute of Biomedicine/Anatomy, University of Helsinki, Helsinki, FIN-00014 Finland
| | - Tarja Porkka-Heiskanen
- Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, FIN-00014 Finland and
| |
Collapse
|
20
|
Hawkins JM, Dubé P, Maloney MT, Wei L, Ewing M, Chesnut SM, Denette JR, Lillie BM, Vaidyanathan R. Synthesis of an H3 Antagonist via Sequential One-Pot Additions of a Magnesium Ate Complex and an Amine to a 1,4-Ketoester followed by Carbonyl-Directed Fluoride Addition. Org Process Res Dev 2012. [DOI: 10.1021/op300093j] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Joel M. Hawkins
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Pascal Dubé
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Mark T. Maloney
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Lulin Wei
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Marcus Ewing
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Stephen M. Chesnut
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Joshua R. Denette
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Brett M. Lillie
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| | - Rajappa Vaidyanathan
- Pharmaceutical Sciences, Pfizer Inc., Eastern Point Road, Groton, Connecticut
06340, United States
| |
Collapse
|
21
|
Samotaeva IS, Birioukova LM, Midzyanovskaya IS, Kuznetsova GD, Bazyan AS, Tuomisto L. Metoprine induced behavioral modifications and brain regional histamine increase in WAG/Rij and Wistar rats. Epilepsy Res 2012; 101:148-56. [DOI: 10.1016/j.eplepsyres.2012.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 03/05/2012] [Accepted: 03/19/2012] [Indexed: 11/26/2022]
|
22
|
Chen CR, Zhou XZ, Luo YJ, Huang ZL, Urade Y, Qu WM. Magnolol, a major bioactive constituent of the bark of Magnolia officinalis, induces sleep via the benzodiazepine site of GABA(A) receptor in mice. Neuropharmacology 2012; 63:1191-9. [PMID: 22771461 DOI: 10.1016/j.neuropharm.2012.06.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 01/12/2023]
Abstract
Magnolol (6,6',7,12-tetramethoxy-2,2'-dimethyl-1-beta-berbaman, C(18)H(18)O(2)), an active ingredient of the bark of Magnolia officinalis, has been reported to exert potent anti-epileptic effects via the GABA(A) receptor. The receptor also mediates sleep in humans and animals. The aim of this study was to determine whether magnolol could modulate sleep behaviors by recording EEG and electromyogram in mice. The results showed that magnolol administered i.p. at a dose of 5 or 25 mg/kg could significantly shorten the sleep latency, increase the amount of non-rapid eye movement (non-REM, NREM) and rapid eye movement (REM) sleep for 3 h after administration with an increase in the number of NREM and REM sleep episodes. Magnolol at doses of 5 and 25 mg/kg increased the number of bouts of wakefulness but decreased their duration. On the other hand, magnolol increased the number of state transitions from wakefulness to NREM sleep and subsequently from NREM sleep to wakefulness. Immunohistochemical study showed that magnolol increased c-Fos expression in the neurons of ventrolateral preoptic area, a sleep center in the anterior hypothalamus, and decreased c-Fos expression in the arousal tuberomammillary nucleus, which was located in the caudolateral hypothalamus. The sleep-promoting effects and changes in c-Fos induced by magnolol were reversed by flumazenil, an antagonist at the benzodiazepine site of the GABA(A) receptor. These results indicate that magnolol increased NREM and REM sleep via the GABA(A) receptor.
Collapse
Affiliation(s)
- Chang-Rui Chen
- Department of Pharmacology, Fudan University, Shanghai, 200032, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Zhao P, Shao Y, Zhang M, Fan K, Kong X, Wang R, Hou Y. Neuropeptide S promotes wakefulness through activation of the posterior hypothalamic histaminergic and orexinergic neurons. Neuroscience 2012; 207:218-26. [DOI: 10.1016/j.neuroscience.2012.01.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Revised: 11/29/2011] [Accepted: 01/09/2012] [Indexed: 10/14/2022]
|
24
|
Hypothalamic Control of Sleep in Aging. Neuromolecular Med 2012; 14:139-53. [DOI: 10.1007/s12017-012-8175-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 02/10/2012] [Indexed: 12/23/2022]
|
25
|
Rao AU, Shao N, Aslanian RG, Chan TY, Degrado SJ, Wang L, McKittrick B, Senior M, West RE, Williams SM, Wu RL, Hwa J, Patel B, Zheng S, Sondey C, Palani A. Discovery of a potent thiadiazole class of histamine h3 receptor antagonist for the treatment of diabetes. ACS Med Chem Lett 2012; 3:198-202. [PMID: 24900450 DOI: 10.1021/ml200250t] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 11/20/2011] [Indexed: 12/31/2022] Open
Abstract
A series of novel 2-piperidinopiperidine thiadiazoles were synthesized and evaluated as new leads of histamine H3 receptor antagonists. The 4-(5-([1,4'-bipiperidin]-1'-yl)-1,3,4-thiadiazol-2-yl)-2-(pyridin-2-yl)morpholine (5u) displayed excellent potency and ex vivo receptor occupancy. Compound 5u was also evaluated in vivo for antidiabetic efficacy in STZ diet-induced obesity type 2 diabetic mice for 2 or 12 days. Non-fasting glucose levels were significantly reduced as compared with vehicle-treated mice. In addition, 5u dose dependently blocked the increase of HbA1c after 12 days of treatment.
Collapse
Affiliation(s)
- Ashwin U. Rao
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Ning Shao
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Robert G. Aslanian
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Tin-Yau Chan
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Sylvia J. Degrado
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Li Wang
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Brian McKittrick
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Mary Senior
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Robert E. West
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Shirley M. Williams
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Ren-Long Wu
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Joyce Hwa
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Bhuneshwari Patel
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Shuqin Zheng
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Christopher Sondey
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| | - Anandan Palani
- Department
of Medicinal Chemistry and ‡Cardiovascular/Metabolic Disease, Merck Research Laboratories, 2015 Galloping Hill Road,
Kenilworth, New Jersey 07033, United States
| |
Collapse
|
26
|
Synthesis and structure–activity relationship (SAR) study of 4-azabenzoxazole analogues as H3 antagonists. Bioorg Med Chem Lett 2012; 22:2075-8. [DOI: 10.1016/j.bmcl.2012.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 11/24/2022]
|
27
|
Haas HL, Lin JS. Waking with the hypothalamus. Pflugers Arch 2011; 463:31-42. [DOI: 10.1007/s00424-011-0996-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/05/2011] [Accepted: 07/12/2011] [Indexed: 12/25/2022]
|
28
|
Wallace TL, Ballard TM, Pouzet B, Riedel WJ, Wettstein JG. Drug targets for cognitive enhancement in neuropsychiatric disorders. Pharmacol Biochem Behav 2011; 99:130-45. [PMID: 21463652 DOI: 10.1016/j.pbb.2011.03.022] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 03/18/2011] [Accepted: 03/28/2011] [Indexed: 12/11/2022]
Abstract
The investigation of novel drug targets for treating cognitive impairments associated with neurological and psychiatric disorders remains a primary focus of study in central nervous system (CNS) research. Many promising new therapies are progressing through preclinical and clinical development, and offer the potential of improved treatment options for neurodegenerative diseases such as Alzheimer's disease (AD) as well as other disorders that have not been particularly well treated to date like the cognitive impairments associated with schizophrenia (CIAS). Among targets under investigation, cholinergic receptors have received much attention with several nicotinic agonists (α7 and α4β2) actively in clinical trials for the treatment of AD, CIAS and attention deficit hyperactivity disorder (ADHD). Both glutamatergic and serotonergic (5-HT) agonists and antagonists have profound effects on neurotransmission and improve cognitive function in preclinical experiments with animals; some of these compounds are now in proof-of-concept studies in humans. Several histamine H3 receptor antagonists are in clinical development not only for cognitive enhancement, but also for the treatment of narcolepsy and cognitive deficits due to sleep deprivation because of their expression in brain sleep centers. Compounds that dampen inhibitory tone (e.g., GABA(A) α5 inverse agonists) or elevate excitatory tone (e.g., glycine transporter inhibitors) offer novel approaches for treating diseases such as schizophrenia, AD and Down syndrome. In addition to cell surface receptors, intracellular drug targets such as the phosphodiesterases (PDEs) are known to impact signaling pathways that affect long-term memory formation and working memory. Overall, there is a genuine need to treat cognitive deficits associated with many neuropsychiatric conditions as well as an increasingly aging population.
Collapse
MESH Headings
- Animals
- Cognition Disorders/drug therapy
- Cognition Disorders/physiopathology
- Glycine Plasma Membrane Transport Proteins/drug effects
- Glycine Plasma Membrane Transport Proteins/physiology
- Humans
- Learning/drug effects
- Learning/physiology
- Memory/drug effects
- Memory/physiology
- Nootropic Agents/pharmacology
- Phosphodiesterase Inhibitors/pharmacology
- Receptors, Cholinergic/drug effects
- Receptors, Cholinergic/physiology
- Receptors, Dopamine/drug effects
- Receptors, Dopamine/physiology
- Receptors, GABA/drug effects
- Receptors, GABA/physiology
- Receptors, Glutamate/drug effects
- Receptors, Glutamate/physiology
- Receptors, Histamine/drug effects
- Receptors, Histamine/physiology
- Receptors, Serotonin/drug effects
- Receptors, Serotonin/physiology
Collapse
Affiliation(s)
- Tanya L Wallace
- Center for Neuroscience, SRI International, Menlo Park, CA, USA
| | | | | | | | | |
Collapse
|
29
|
Lin JS, Anaclet C, Sergeeva OA, Haas HL. The waking brain: an update. Cell Mol Life Sci 2011; 68:2499-512. [PMID: 21318261 PMCID: PMC3134769 DOI: 10.1007/s00018-011-0631-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Revised: 12/25/2010] [Accepted: 01/13/2011] [Indexed: 11/01/2022]
Abstract
Wakefulness and consciousness depend on perturbation of the cortical soliloquy. Ascending activation of the cerebral cortex is characteristic for both waking and paradoxical (REM) sleep. These evolutionary conserved activating systems build a network in the brainstem, midbrain, and diencephalon that contains the neurotransmitters and neuromodulators glutamate, histamine, acetylcholine, the catecholamines, serotonin, and some neuropeptides orchestrating the different behavioral states. Inhibition of these waking systems by GABAergic neurons allows sleep. Over the past decades, a prominent role became evident for the histaminergic and the orexinergic neurons as a hypothalamic waking center.
Collapse
Affiliation(s)
- Jian-Sheng Lin
- INSERM-U628, Integrative Physiology of Brain Arousal Systems, Claude Bernard University, 69373, Lyon, France
| | | | | | | |
Collapse
|
30
|
Abstract
The histaminergic system fulfills a major role in the maintenance of waking. Histaminergic neurons are located exclusively in the posterior hypothalamus from where they project to most areas of the central nervous system. The histamine H(3) receptors are autoreceptors damping histamine synthesis, the firing frequency of histamine neurons, and the release of histamine from axonal varicosities. It is noteworthy that this action also extends to heteroreceptors on the axons of most other neurotransmitter systems, allowing a powerful control over multiple homeostatic functions. The particular properties and locations of histamine H(3) receptors provide quite favorable attributes to make this a most promising target for pharmacological interventions of sleep and waking disorders associated with narcolepsy, Parkinson's disease, and other neuropsychiatric indications.
Collapse
Affiliation(s)
- Jian-Sheng Lin
- Institut National de la Santé et de la Recherche Médicale, Integrative Physiology of Brain Arousal Systems, Claude Bernard University, Lyon, France
| | | | | |
Collapse
|
31
|
Sakai K, Takahashi K, Anaclet C, Lin JS. Sleep-waking discharge of ventral tuberomammillary neurons in wild-type and histidine decarboxylase knock-out mice. Front Behav Neurosci 2010; 4:53. [PMID: 21060718 PMCID: PMC2972729 DOI: 10.3389/fnbeh.2010.00053] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 07/21/2010] [Indexed: 01/30/2023] Open
Abstract
Using extracellular single-unit recordings, we have determined the characteristics of neurons in the ventral tuberomammillary nucleus (VTM) of wild-type (WT) and histidine decarboxylase knock-out (HDC-KO) mice during the sleep-waking cycle. The VTM neurons of HDC-KO mice showed no histamine immunoreactivity, but were immunoreactive for the histaminergic (HA) neuron markers adenosine deaminase and glutamic acid decarboxylase 67. In the VTM of WT mice, we found waking (W)-specific, non-W-specific W-active, sleep-active, W and paradoxical sleep (PS)-active, and state-indifferent neuron groups. We previously demonstrated in WT mice that only W-specific neurons are histaminergic and that they are characterized by a triphasic broad action potential. In the VTM of HDC-KO mice, we found all these groups of state-dependent and state-indifferent neurons, including W-specific neurons that were characterized by a triphasic broad action potential and a W-specific slow tonic discharge, as in WT mice. The W-specific neurons ceased firing before the onset of electroencephalogram (EEG) synchronization, the first EEG sign of sleep, and remained silent during both slow-wave sleep (SWS) and PS. At the transition from SWS to W, they discharged after the onset of EEG activation, the first EEG sign of W. They either responded to an arousing stimulus with a long delay or did not respond. They therefore presented exactly the same characteristics as those seen in the VTM of WT mice. Thus VTM neurons deprived of their natural transmitter histamine still exhibit the firing properties of W-specific HA neurons.
Collapse
Affiliation(s)
- Kazuya Sakai
- INSERM U628, Claude Bernard University Lyon 1 Lyon, France
| | | | | | | |
Collapse
|
32
|
Abstract
Orexin neurons (hypocretin neurons) have a critical role in the regulation of sleep/wakefulness, especially in the maintenance of arousal. Here, we revealed that orexin neurons are directly and indirectly activated by orexin via the orexin 2 receptor (OX2R). Orexin B (1 μM) induced depolarization in orexin neurons, which was still observed in the presence of TTX (1 μM), AP-5 (50 μM), and CNQX (20 μM). In addition, orexin B induced inward currents in the presence of TTX, suggesting a direct activation of orexin neurons. Although orexin B application induced depolarization in orexin neurons of OX1R knock-out mice at comparable levels to wild-type mice, the observation that orexin B failed to depolarize orexin neurons in the OX2R knock-out mice suggested that OX2R was a primary receptor for this response. Moreover, immunoelectron microscopic analyses revealed direct contacts among orexin neurons, which exhibited structural similarities to the glutamatergic synapses. Together, these results suggest that orexin neurons form a positive-feedback circuit through indirect and direct pathways, which results in the preservation of the orexin neuron network at a high activity level and/or for a longer period. Therefore, the activation of orexin neurons through OX2R might have an important role in the maintenance of arousal.
Collapse
|
33
|
Thakkar MM. Histamine in the regulation of wakefulness. Sleep Med Rev 2010; 15:65-74. [PMID: 20851648 DOI: 10.1016/j.smrv.2010.06.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 06/13/2010] [Accepted: 06/15/2010] [Indexed: 11/26/2022]
Abstract
The histaminergic system is exclusively localized within the posterior hypothalamus with projection to almost all the major regions of the central nervous system. Strong and consistent evidence exist to suggest that histamine, acting via H₁ and/or H₃ receptor has a pivotal role in the regulation of sleep-wakefulness. Administration of histamine or H₁ receptor agonists induces wakefulness, whereas administration of H₁ receptor antagonists promotes sleep. The H₃ receptor functions as an auto-receptor and regulates the synthesis and release of histamine. Activation of H₃ receptor reduces histamine release and promotes sleep. Conversely, blockade of H₃ receptor promotes wakefulness. Histamine release in the hypothalamus and other target regions is highest during wakefulness. The histaminergic neurons display maximal activity during the state of high vigilance, and cease their activity during non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. The cerebrospinal levels of histamine are reduced in diseased states where hypersomnolence is a major symptom. The histamine deficient L-histidine decarboxylase knockout (HDC KO) mice display sleep fragmentation and increased REM sleep during the light period along with profound wakefulness deficit at dark onset, and in novel environment. Similar results have been obtained when histamine neurons are lesioned. These studies strongly implicate the histaminergic neurons of the TMN to play a critical role in the maintenance of high vigilance state during wakefulness.
Collapse
Affiliation(s)
- Mahesh M Thakkar
- Neurology, University of Missouri, Harry S. Truman Memorial Veterans Hospital, Research, Room A023, 800 Hospital Drive, Columbia, MO 65210, USA.
| |
Collapse
|
34
|
Van Ruitenbeek P, Vermeeren A, Riedel WJ. Cognitive domains affected by histamine H(1)-antagonism in humans: a literature review. ACTA ACUST UNITED AC 2010; 64:263-82. [PMID: 20685608 DOI: 10.1016/j.brainresrev.2010.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 04/23/2010] [Accepted: 04/24/2010] [Indexed: 11/18/2022]
Abstract
The neurotransmitter histamine has been suggested to be involved in cognitive functioning. Generally, studies in animals have shown a decrease in performance after decreasing histamine neurotransmission and improved performance after increasing histamine neurotransmission. It is unclear, however, what role histamine plays in cognition in humans. Up until now, most data are derived from studies and reviews that aimed to assess the sedative potential of H(1)-antagonists and not the effects on cognition in particular. The objective of this paper is specifically to review which cognitive domains are affected by H(1)-antagonists. Taken together, 90 experimental studies on the performance effects of sedative H(1)-antagonists published between 1973 and 2009 were reviewed. Results showed that psychomotor skills and attention are most frequently impaired and memory the least. Tasks assessing memory that were affected usually required rapid responses. It was concluded that both the complexity of the task as well as the demand for information processing speed determines the sensitivity to the effects of central H(1)-antagonism. The importance of the sensitive cognitive domains to histaminergic dysfunction, as well as the relation between histamine related decrease in arousal and task performance deserve further research.
Collapse
Affiliation(s)
- P Van Ruitenbeek
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, The Netherlands, Maastricht, The Netherlands.
| | | | | |
Collapse
|
35
|
Church MK, Maurer M, Simons FER, Bindslev-Jensen C, van Cauwenberge P, Bousquet J, Holgate ST, Zuberbier T. Risk of first-generation H(1)-antihistamines: a GA(2)LEN position paper. Allergy 2010; 65:459-66. [PMID: 20146728 DOI: 10.1111/j.1398-9995.2009.02325.x] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND First-generation H(1)-antihistamines obtained without prescription are the most frequent form of self-medication for allergic diseases, coughs and colds and insomnia even though they have potentially dangerous unwanted effects which are not recognized by the general public. AIMS To increase consumer protection by bringing to the attention of regulatory authorities, physicians and the general public the potential dangers of the indiscriminate use first-generation H(1)-antihistamines purchased over-the counter in the absence of appropriate medical supervision. METHODS A GA(2)LEN (Global Allergy and Asthma European Network) task force assessed the unwanted side-effects and potential dangers of first-generation H1-antihistamines by reviewing the literature (Medline and Embase) and performing a media audit of US coverage from 1996 to 2008 of accidents and fatal adverse events in which these drugs were implicated. RESULTS First-generation H(1)-antihistamines, all of which are sedating, are generally regarded as safe by laypersons and healthcare professionals because of their long-standing use. However, they reduce rapid eye movement (REM)-sleep, impair learning and reduce work efficiency. They are implicated in civil aviation, motor vehicle and boating accidents, deaths as a result of accidental or intentional overdosing in infants and young children and suicide in teenagers and adults. Some exhibit cardiotoxicity in overdose. CONCLUSIONS This review raises the issue of better consumer protection by recommending that older first-generation H(1)-antihistamines should no longer be available over-the-counter as prescription- free drugs for self-medication of allergic and other diseases now that newer second- generation nonsedating H(1)-antihistamines with superior risk/benefit ratios are widely available at competitive prices.
Collapse
Affiliation(s)
- M K Church
- Department of Dermatology and Allergy, Charité- Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hondo M, Nagai K, Ohno K, Kisanuki Y, Willie JT, Watanabe T, Yanagisawa M, Sakurai T. Histamine-1 receptor is not required as a downstream effector of orexin-2 receptor in maintenance of basal sleep/wake states. Acta Physiol (Oxf) 2010; 198:287-94. [PMID: 19694625 PMCID: PMC3513392 DOI: 10.1111/j.1748-1716.2009.02032.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AIM The effect of orexin on wakefulness has been suggested to be largely mediated by activation of histaminergic neurones in the tuberomammillary nucleus (TMN) via orexin receptor-2 (OX(2)R). However, orexin receptors in other regions of the brain might also play important roles in maintenance of wakefulness. To dissect the role of the histaminergic system as a downstream mediator of the orexin system in the regulation of sleep/wake states without compensation by the orexin receptor-1 (OX(1)R) mediated pathways, we analysed the phenotype of Histamine-1 receptor (H(1)R) and OX(1)R double-deficient (H(1)R(-/-);OX(1)R(-/-)) mice. These mice lack OX(1)R-mediated pathways in addition to deficiency of H(1)R, which is thought to be the most important system in downstream of OX(2)R. METHODS We used H(1)R deficient (H(1)R(-/-)) mice, H(1)R(-/-);OX(1)R(-/-) mice, OX(1)R and OX(2)R double-deficient (OX(1)R(-/-);OX(2)R(-/-)) mice, and wild type controls. Rapid eye movement (REM) sleep, non-REM (NREM) sleep and awake states were determined by polygraphic electroencephalographic/electromyographic recording. RESULTS No abnormality in sleep/wake states was observed in H(1)R(-/-) mice, consistent with previous studies. H(1)R(-/-);OX(1)R(-/-) mice also showed a sleep/wake phenotype comparable to that of wild type mice, while OX(1)R(-/-); OX(2)R(-/-) mice showed severe fragmentation of sleep/wake states. CONCLUSION Our observations showed that regulation of the sleep/wake states is completely achieved by OX(2)R-expressing neurones without involving H(1)R-mediated pathways. The maintenance of basal physiological sleep/wake states is fully achieved without both H(1) and OX(1) receptors. Downstream pathways of OX(2)R other than the histaminergic system might play an important role in the maintenance of sleep/wake states.
Collapse
MESH Headings
- Animals
- Antigens, Surface/metabolism
- Brain/physiology
- Electroencephalography
- Electromyography
- Male
- Mice
- Mice, Knockout
- Neurons/physiology
- Orexin Receptors
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Histamine H1/deficiency
- Receptors, Histamine H1/metabolism
- Receptors, Neuropeptide/deficiency
- Receptors, Neuropeptide/metabolism
- Sleep/physiology
- Sleep, REM/physiology
- Wakefulness/physiology
Collapse
Affiliation(s)
- M. Hondo
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Pharmacology, Institute of Basic Medical Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - K. Nagai
- Department of Pharmacology, Institute of Basic Medical Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - K. Ohno
- Department of Pharmacology, Institute of Basic Medical Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Y. Kisanuki
- Department of Molecular Genetics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - J. T. Willie
- Department of Molecular Genetics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - T. Watanabe
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - M. Yanagisawa
- Department of Molecular Genetics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - T. Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Pharmacology, Institute of Basic Medical Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
37
|
Denonne F, Atienzar F, Célanire S, Christophe B, Delannois F, Delaunoy C, Delporte ML, Durieu V, Gillard M, Lallemand B, Lamberty Y, Lorent G, Vanbellinghen A, Van houtvin N, Verbois V, Provins L. Phenyl-oxazoles, a New Family of Inverse Agonists at the H3Histamine Receptor. ChemMedChem 2010; 5:206-12. [DOI: 10.1002/cmdc.200900446] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Orexin/hypocretin and histamine: distinct roles in the control of wakefulness demonstrated using knock-out mouse models. J Neurosci 2009; 29:14423-38. [PMID: 19923277 DOI: 10.1523/jneurosci.2604-09.2009] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To determine the respective role played by orexin/hypocretin and histamine (HA) neurons in maintaining wakefulness (W), we characterized the behavioral and sleep-wake phenotypes of orexin (Ox) knock-out (-/-) mice and compared them with those of histidine-decarboxylase (HDC, HA-synthesizing enzyme)-/- mice. While both mouse strains displayed sleep fragmentation and increased paradoxical sleep (PS), they presented a number of marked differences: (1) the PS increase in HDC(-/-) mice was seen during lightness, whereas that in Ox(-/-) mice occurred during darkness; (2) contrary to HDC(-/-), Ox(-/-) mice had no W deficiency around lights-off, nor an abnormal EEG and responded to a new environment with increased W; (3) only Ox(-/-), but not HDC(-/-) mice, displayed narcolepsy and deficient W when faced with motor challenge. Thus, when placed on a wheel, wild-type (WT), but not littermate Ox(-/-) mice, voluntarily spent their time in turning it and as a result, remained highly awake; this was accompanied by dense c-fos expression in many areas of their brains, including Ox neurons in the dorsolateral hypothalamus. The W and motor deficiency of Ox(-/-) mice was due to the absence of Ox because intraventricular dosing of orexin-A restored their W amount and motor performance whereas SB-334867 (Ox1-receptor antagonist, i.p.) impaired W and locomotion of WT mice during the test. These data indicate that Ox, but not HA, promotes W through enhanced locomotion and suggest that HA and Ox neurons exert a distinct, but complementary and synergistic control of W: the neuropeptide being more involved in its behavioral aspects, whereas the amine is mainly responsible for its qualitative cognitive aspects and cortical EEG activation.
Collapse
|
39
|
Rao AU, Palani A, Chen X, Huang Y, Aslanian RG, West RE, Williams SM, Wu RL, Hwa J, Sondey C, Lachowicz J. Synthesis and structure-activity relationships of 2-(1,4'-bipiperidin-1'-yl)thiazolopyridine as H3 receptor antagonists. Bioorg Med Chem Lett 2009; 19:6176-80. [PMID: 19773164 DOI: 10.1016/j.bmcl.2009.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 08/31/2009] [Accepted: 09/02/2009] [Indexed: 11/28/2022]
Abstract
A series of 2-(1,4'-bipiperidine-1'-yl)thiazolopyridines was synthesized and evaluated as a new lead of non-imidazole histamine H(3) receptor antagonists. Introduction of diversity at the 6-position of the pyridine ring was designed to enhance in vitro potency and decrease hERG activity. The structure-activity relationships for these new thiazolopyridine antagonists are discussed.
Collapse
Affiliation(s)
- Ashwin U Rao
- Schering-Plough Research Institute, 2015 Galloping Hill Road, K-15-1-1800, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhao C, Sun M, Bennani YL, Miller TR, Witte DG, Esbenshade TA, Wetter J, Marsh KC, Hancock AA, Brioni JD, Cowart MD. Design of a New Histamine H3 Receptor Antagonist Chemotype: (3aR,6aR)-5-Alkyl-1-aryl-octahydropyrrolo[3,4-b]pyrroles, Synthesis, and Structure−Activity Relationships. J Med Chem 2009; 52:4640-9. [DOI: 10.1021/jm900480x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chen Zhao
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Minghua Sun
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Youssef L. Bennani
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Thomas R. Miller
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - David G. Witte
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Timothy A. Esbenshade
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Jill Wetter
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Kennan C. Marsh
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Arthur A. Hancock
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Jorge D. Brioni
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Marlon D. Cowart
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| |
Collapse
|
41
|
Zhao C, Sun M, Bennani YL, Gopalakrishnan SM, Witte DG, Miller TR, Krueger KM, Browman KE, Thiffault C, Wetter J, Marsh KC, Hancock AA, Esbenshade TA, Cowart MD. The Alkaloid Conessine and Analogues as Potent Histamine H3 Receptor Antagonists. J Med Chem 2008; 51:5423-30. [DOI: 10.1021/jm8003625] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chen Zhao
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Minghua Sun
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Youssef L. Bennani
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Sujatha M. Gopalakrishnan
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - David G. Witte
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Thomas R. Miller
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Kathleen M. Krueger
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Kaitlin E. Browman
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Christine Thiffault
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Jill Wetter
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Kennan C. Marsh
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Arthur A. Hancock
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Timothy A. Esbenshade
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| | - Marlon D. Cowart
- Department of Neuroscience Research, Global Pharmaceutical Research Division, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6123
| |
Collapse
|
42
|
Le S, Gruner JA, Mathiasen JR, Marino MJ, Schaffhauser H. Correlation between ex vivo receptor occupancy and wake-promoting activity of selective H3 receptor antagonists. J Pharmacol Exp Ther 2008; 325:902-9. [PMID: 18305012 DOI: 10.1124/jpet.107.135343] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
The histamine H3 receptor (H3R) modulates the release of neurotransmitters that are involved in vigilance, cognition, and sleep-wake regulation. H3R antagonism has been proposed as a novel approach to the treatment of cognitive and attention deficit as well as sleep disorders. It is apparent that H3R antagonists produce pharmacological effects in preclinical animal models across a wide dose range. Several H3R antagonists were reported to be effective at producing cognitive enhancing effects at low doses, while producing robust wake enhancement at higher doses. To better understand the effect of H3R antagonists across a broad dose range, an ex vivo receptor binding assay has been used to estimate the degree of H3R occupancy in vivo. The H3R antagonists ciproxifan, thioperamide, GSK189254 (6-[(3-cyclobutyl-2,3,4,5-tetrahydro-1H-3-benzazepin-7-yl)oxy]-N-methyl-3-pyridinecarboxamide hydrochloride), and ABT-239 ([4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile) produced wake-promoting activity in vivo and a dose-dependent inhibition of H3R binding ex vivo. For ciproxifan, thioperamide, and GSK189254, a relatively low level of cumulative wake activity was linearly correlated with up to 80% of the receptor occupancy. In contrast, an abrupt break from linearity and a robust increase of waking activity was observed at doses that produce greater than 80% occupancy. Our results suggest a relatively small increase of waking activity at low levels of receptor occupancy that may be consistent with reported enhancement of attention and cognitive function. Robust waking activity at higher levels of H3R occupancy may be mechanistically different from activities at low levels of H3R occupancy.
Collapse
Affiliation(s)
- Siyuan Le
- World Wide Discovery Research, CNS Biology, Cephalon, Inc., 145 Brandywine Parkway, West Chester, PA 19380, USA.
| | | | | | | | | |
Collapse
|
43
|
Fiske E, Portas CM, Grønli J, Sørensen E, Bjorvatn B, Bjørkum AA, Ursin R. Increased extracellular 5-HT but no change in sleep after perfusion of a 5-HT1A antagonist into the dorsal raphe nucleus of rats. Acta Physiol (Oxf) 2008; 193:89-97. [PMID: 18081887 DOI: 10.1111/j.1748-1716.2007.01792.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM The 5-HT(1A) receptor antagonist 4-Iodo-N-[2-[4-(methoxyphenyl)-1-piperazinyl]ethyl]-N-2-pyridinyl-benzamide hydrochloride (p-MPPI) (10 microM) was perfused into the dorsal raphe nucleus (DRN) to study simultaneously the effects of the drug on the DRN and frontal cortex extracellular serotonin (5-hydroxytryptamine, 5-HT) levels and concurring behavioural states. METHODS Waking, slow wave sleep and rapid eye movement sleep were determined by polygraphic recordings during microdialysis perfusion and extracellular sample collection. The samples were analysed by microbore high-performance liquid chromatography coupled with electrochemical detection for analysis of 5-HT. RESULTS p-MPPI perfusion into the DRN (n = 6) produced a sixfold 5-HT increase in the DRN during all behavioural states. The increased 5-HT level was most likely related to the blockage of 5-HT(1A) receptors in the DRN by p-MPPI. No significant effect was seen on sleep. CONCLUSION Despite the dramatic increase in DRN extracellular 5-HT produced by p-MPPI, only a transient and nonsignificant effect on sleep was recorded. It is suggested that the usual coupling between 5-HT level and behavioural state may be lost when an excessive serotonergic output is pharmacologically achieved.
Collapse
Affiliation(s)
- E Fiske
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | | | | | | | | | | | | |
Collapse
|
44
|
Correa JV, Herrera B, Toro-Labbé A. Characterization of the reactive conformations of protonated histamine through the reaction force analysis and the dual descriptor of chemical reactivity. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.theochem.2007.04.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
45
|
Isensee K, Petroianu G, Stark H. Pharmacological aspects of cognitive impairment: past, present and future of drugs in dementia. J Appl Biomed 2007. [DOI: 10.32725/jab.2007.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
46
|
Medalha CC, Mattioli R. Involvement of the histaminergic system on appetitive learning and its interaction with haloperidol in goldfish. Neurosci Lett 2007; 418:195-200. [PMID: 17386974 DOI: 10.1016/j.neulet.2007.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 02/26/2007] [Accepted: 03/09/2007] [Indexed: 11/24/2022]
Abstract
This study investigated the actions of the histaminergic system on appetitive learning and memory, and its interaction with the dopaminergic system in goldfish. It consisted of nine sessions, in which fish were tested in a four-arm tank. On day 1, the animals were habituated for 10 min. On day 2, they were placed in one arm and had to find food at the left or the right arm. Time to begin feeding was recorded, and the procedure repeated for more 3 days (training phase). On training day 4, seven groups were injected with saline, seven with haloperidol (2.0 mg/kg) and one with DMSO solution before training and after feeding, three groups received saline, six chlorpheniramine (CPA) (1.0, 4.0 and 8.0 mg/kg), and six l-histidine (LH) (25, 50 and 100 mg/kg). Saline groups were considered as control of CPA and LH treated groups and DMSO as control of haloperidol. A non-injected group was also included. Testing occurred after 24 h. A reversal procedure was conducted 24h after testing and repeated for 3 days. The groups receiving CPA at 1.0 and 8.0 mg/kg and LH at 25, 50 and 100 mg/kg differed between Test and Reversal day 1. Pre-treatment with haloperidol plus 8.0 mg/kg of CPA and 25 and 50 mg/kg of LH reverted the treatment effect. However, in the groups treated with 1.0 mg/kg of CPA and 100 mg/kg of LH, the difference remained. This study confirmed the interaction between the histaminergic and the dopaminergic systems on memory process in goldfish.
Collapse
|
47
|
Zhang M, Thurmond RL, Dunford PJ. The histamine H4 receptor: A novel modulator of inflammatory and immune disorders. Pharmacol Ther 2007; 113:594-606. [PMID: 17275092 DOI: 10.1016/j.pharmthera.2006.11.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 11/29/2006] [Accepted: 11/29/2006] [Indexed: 02/01/2023]
Abstract
All 4 known histamine receptors (H(1)R, H(2)R, H(3)R and H(4)R) have been used or proposed as therapeutic targets for varied diseases. This article reviews the recent progress in understanding the function of the recently described histamine receptor H(4)R in a variety of immune responses and the potential therapeutic value of H(4)R antagonists. The H(4)R is expressed primarily on cells involved in inflammation and immune response. It has effects on chemotaxis, as well as cytokine and chemokine production of mast cells, eosinophils, dendritic cells, and T cells. H(4)R antagonists, JNJ 7777120 and JNJ 10191584 (also known as VUF 6002) have been developed with excellent affinity and selectivity towards human and rodent H(4)R. These antagonists also demonstrate efficacy as anti-inflammatory agents in vivo. H(4)R antagonists have shown promising activity in down-regulating immune responses in a range of animal disease models including acute inflammation, hapten-mediated colitis, and allergic airway inflammation. Due to its distribution on immune cells and its proven role in inflammatory functions, the H(4)R appears to be a therapeutic target for the treatment of a variety of immune disorders.
Collapse
Affiliation(s)
- Mai Zhang
- Johnson & Johnson Pharmaceutical Research and Development L.L.C., 3210 Merryfield Row, San Diego, CA 92121, USA
| | | | | |
Collapse
|
48
|
Celanire S, Wijtmans M, Talaga P, Leurs R, de Esch IJP. Keynote review: histamine H3 receptor antagonists reach out for the clinic. Drug Discov Today 2006; 10:1613-27. [PMID: 16376822 DOI: 10.1016/s1359-6446(05)03625-1] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Antagonists of the histamine H(1) and H(2) receptors have been successful as blockbuster drugs for treating allergic conditions and gastric ulcers, respectively. As such, histamine receptors have made a significant contribution to establishing G-Protein-coupled receptors as the favored drug targets of the industry. In this light, it can easily be understood that the discovery of a third histamine receptor subtype (H(3)R) in 1983 was greeted with considerable excitement. However, characterization of the H(3)R turned out to be far from trivial. In the past five years, molecular biology approaches have given fresh impetus to the H(3)R research field. As a result, H(3)R ligands are where they were anticipated to be 20 years ago: at the center of attention and on the verge of an anticipated breakthrough as the next generation of histaminergic blockbuster drugs. Here, we assess the status of the H(3)R medicinal chemistry programs of the various players in the field, as far as can be deduced from patent applications and scientific literature.
Collapse
Affiliation(s)
- Sylvain Celanire
- Department of Chemical Research, Chemin du Foriest, B-1420 Braine-l'Alleud, Belgium
| | | | | | | | | |
Collapse
|
49
|
Dai H, Okuda T, Sakurai E, Kuramasu A, Kato M, Jia F, Xu AJ, Iinuma K, Sato I, Yanai K. Blockage of histamine H1 receptor attenuates social isolation-induced disruption of prepulse inhibition: a study in H1 receptor gene knockout mice. Psychopharmacology (Berl) 2005; 183:285-93. [PMID: 16237577 DOI: 10.1007/s00213-005-0203-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Accepted: 09/14/2005] [Indexed: 10/25/2022]
Abstract
RATIONALE Histaminergic neurotransmission has been implicated in the pathophysiology of stress-related psychiatric diseases. Although several atypical antipsychotics are potent H1 antagonists, the clinical significance of interaction between atypical antipsychotics and H1 receptors is still unknown. OBJECTIVE In this study, we investigated the effects of H1 receptors blockage on social isolation-induced behavioral changes in H1 receptor gene knockout (H1KO) mice and their wild-type (WT) mice. METHODS Both H1KO and their WT mice were subjected to 4-week social isolation rearing after weaning (21 postnatal days). After the 4-week isolation period, mice behavioral changes were evaluated using behavioral tests. RESULTS Locomotor activity in home cages was significantly lower in isolation-reared WT mice than in socially reared WT mice. However, no change in locomotor activity was observed between socially and isolation-reared H1KO mice. Social isolation significantly impaired prepulse inhibition (PPI) of startle response in WT mice but not in H1KO mice. In addition, social isolation significantly impaired spatial learning and memory in WT mice but not in H1KO mice. Furthermore, H1KO mice treated with methamphetamine (METH) showed no enhancement in isolation-induced disruption of PPI. A neurochemical study revealed that isolation-reared WT mice had significantly lower dopamine (DA) levels and slightly increased DA turnover in the cortex than socially reared WT mice. Conversely, isolation-reared H1KO mice showed significantly higher DA contents as compared with socially reared H1KO mice. CONCLUSION The results of our study indicate that blockage of H1 receptor-mediated neurotransmission attenuates social isolation-induced behavioral changes and that the therapeutic effects of atypical antipsychotics are mediated, at least in part, by interaction with H1 receptors in the brain.
Collapse
Affiliation(s)
- Hongmei Dai
- Department of Pharmacology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, 980-8575 Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
In sleep medicine, patients with sleep disorders are evaluated and treated. The primary assessment tool of the field has traditionally been polysomnography. While polysomnography has been helpful in the evaluation of some sleep disorders, such as sleep apnea syndrome and periodic limb movement disorder, it has been less helpful in others, such as the insomnias, or sleep disorders secondary to mental disorders. These disorders are presumed to stem from some alteration in brain function that disrupts sleep. The development of functional neuroimaging methods provides a means to understand brain function in patients with sleep disorders in a manner not accessible to polysomnography. This paper summarizes functional neuroimaging findings during healthy sleep, then, reviews available studies in sleep disorders patients, and studies addressing the pharmacology of sleep and sleep disorders. Areas in which functional neuroimaging methods may be helpful in sleep medicine, and in which future development is advised, include: (1) clarification of pathophysiology; (2) aid in differential diagnosis; (3) assessment of treatment response; (4) guiding new drug development; and (5) monitoring treatment response.
Collapse
Affiliation(s)
- Eric A Nofzinger
- Sleep Neuroimaging Research Program, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, 3811 O'Hara Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|