1
|
Dremencov E, Jezova D. Editorial: New players on the monoaminergic field: relevance to the mental disorders. Front Pharmacol 2024; 15:1504261. [PMID: 39469628 PMCID: PMC11513357 DOI: 10.3389/fphar.2024.1504261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Affiliation(s)
- Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
2
|
Ma S, Ma Q, Hu S, Mo X, Zhu C, Zhang X, Jia Z, Tang L, Jiang L, Cui Y, Chen Z, Hu W, Zhang X. Deletion of histamine H2 receptor in VTA dopaminergic neurons of mice induces behavior reminiscent of mania. Cell Rep 2024; 43:114717. [PMID: 39264811 DOI: 10.1016/j.celrep.2024.114717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 09/14/2024] Open
Abstract
Hyperfunction of the dopamine system has been implicated in manic episodes in bipolar disorders. How dopaminergic neuronal function is regulated in the pathogenesis of mania remains unclear. Histaminergic neurons project dense efferents into the midbrain dopaminergic nuclei. Here, we present mice lacking dopaminergic histamine H2 receptor (H2R) in the ventral tegmental area (VTA) that exhibit a behavioral phenotype mirroring some of the symptoms of mania, including increased locomotor activity and reduced anxiety- and depression-like behavior. These behavioral deficits can be reversed by the mood stabilizers lithium and valproate. H2R deletion in dopaminergic neurons significantly enhances neuronal activity, concurrent with a decrease in the γ-aminobutyric acid (GABA) type A receptor (GABAAR) membrane presence and inhibitory transmission. Conversely, either overexpression of H2R in VTA dopaminergic neurons or treatment of H2R agonist amthamine within the VTA counteracts amphetamine-induced hyperactivity. Together, our results demonstrate the engagement of H2R in reducing VTA dopaminergic activity, shedding light on the role of H2R as a potential target for mania therapy.
Collapse
Affiliation(s)
- Shijia Ma
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qianyi Ma
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Songhui Hu
- Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China
| | - Xinlei Mo
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Chenze Zhu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xingxian Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zetao Jia
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lingjie Tang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lei Jiang
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yihui Cui
- Department of Neurobiology, Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Weiwei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, Zhejiang 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang 321299, China.
| |
Collapse
|
3
|
Szukiewicz D. Histaminergic System Activity in the Central Nervous System: The Role in Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2024; 25:9859. [PMID: 39337347 PMCID: PMC11432521 DOI: 10.3390/ijms25189859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Histamine (HA), a biogenic monoamine, exerts its pleiotropic effects through four H1R-H4R histamine receptors, which are also expressed in brain tissue. Together with the projections of HA-producing neurons located within the tuberomammillary nucleus (TMN), which innervate most areas of the brain, they constitute the histaminergic system. Thus, while remaining a mediator of the inflammatory reaction and immune system function, HA also acts as a neurotransmitter and a modulator of other neurotransmitter systems in the central nervous system (CNS). Although the detailed causes are still not fully understood, neuroinflammation seems to play a crucial role in the etiopathogenesis of both neurodevelopmental and neurodegenerative (neuropsychiatric) diseases, such as autism spectrum disorders (ASDs), attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD) and Parkinson's disease (PD). Given the increasing prevalence/diagnosis of these disorders and their socioeconomic impact, the need to develop effective forms of therapy has focused researchers' attention on the brain's histaminergic activity and other related signaling pathways. This review presents the current state of knowledge concerning the involvement of HA and the histaminergic system within the CNS in the development of neurodevelopmental and neurodegenerative disorders. To this end, the roles of HA in neurotransmission, neuroinflammation, and neurodevelopment are also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
4
|
Li X, Liang X, Ma S, Zhao S, Wang W, Li M, Feng D, Tang M. SERT and OCT mediate 5-HT 1B receptor regulation of immobility behavior and uptake of 5-HT and HIS. Biomed Pharmacother 2024; 177:117017. [PMID: 38917762 DOI: 10.1016/j.biopha.2024.117017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
5-HT clearance, commonly mediated by transporters in the uptake-1 and uptake-2 families, has been linked to 5-HT1B receptor's action on behaviors. Since no specific transporters identified yet, effects of serotonin transporter (SERT) and organic cation transporter (OCTs) on 5-HT1B-elicited immobility phenotype, and 5-HT and HIS uptake were then investigated. Intraperitoneal injections of SERT inhibitor fluoxetine (FLX) and/or OCTs inhibitor decynium (D22) were used prior to local perfusion of 5-HT1B agonist CP93129 into the ventral hippocampus to measure immobility times in the FST and TST, to measure 5-HT uptake efficiencies and HIS uptake efficiencies derived from linear regressions using the transient no-net-flux quantitative microdialysis in C57BL/6 mice. Exogenous 5-HT and HIS uptake were measured following incubation of FLX and/or D22 with CP93129 in the RBL-2H3 cells. Moreover, surface membrane levels of SERT and OCT were detected in response to CP93129. Local CP93129 prolonged immobility times, which were attenuated following pretreatment of either inhibitor. Local CP93129 lowered the slopes obtained from the lineal regressions for 5-HT and HIS (slope is reciprocal to uptake efficiency), which were then weakened following pretreatment of either inhibitor. Similar findings were obtained following CP93129 incubation, and co-incubation of CP93129 with either inhibitor in the RBL-2H3. Moreover, CP93129 dose-dependently moved SERT and OCT3 in the cytosol to the surface membrane. Both SERT and OCT are the target effectors mediating 5-HT1B regulation of immobility time and 5-HT uptake, OCT mediates 5-HT1B regulation of HIS uptake. Their underlying signal transductions need to be further explored.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xuankai Liang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Shenglu Ma
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Shulei Zhao
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Wenyao Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Mingxing Li
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Dan Feng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Man Tang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
5
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2024:10.1007/s12035-024-04333-y. [PMID: 39012443 DOI: 10.1007/s12035-024-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
6
|
Fu Z, Yang X, Jiang Y, Mao X, Liu H, Yang Y, Chen J, Chen Z, Li H, Zhang XS, Mao X, Li N, Wang D, Jiang J. Microbiota profiling reveals alteration of gut microbial neurotransmitters in a mouse model of autism-associated 16p11.2 microduplication. Front Microbiol 2024; 15:1331130. [PMID: 38596370 PMCID: PMC11002229 DOI: 10.3389/fmicb.2024.1331130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
The gut-brain axis is evident in modulating neuropsychiatric diseases including autism spectrum disorder (ASD). Chromosomal 16p11.2 microduplication 16p11.2dp/+ is among the most prevalent genetic copy number variations (CNV) linked with ASD. However, the implications of gut microbiota status underlying the development of ASD-like impairments induced by 16p11.2dp/+ remains unclear. To address this, we initially investigated a mouse model of 16p11.2dp/+, which exhibits social novelty deficit and repetitive behavior characteristic of ASD. Subsequently, we conducted a comparative analysis of the gut microbial community and metabolomic profiles between 16p11.2dp/+ and their wild-type counterparts using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC/MS). Our microbiota analysis revealed structural dysbiosis in 16p11.2dp/+ mice, characterized by reduced biodiversity and alterations in species abundance, as indicated by α/β-diversity analysis. Specifically, we observed reduced relative abundances of Faecalibaculum and Romboutsia, accompanied by an increase in Turicibacter and Prevotellaceae UCG_001 in 16p11.2dp/+ group. Metabolomic analysis identified 19 significantly altered metabolites and unveiled enriched amino acid metabolism pathways. Notably, a disruption in the predominantly histamine-centered neurotransmitter network was observed in 16p11.2dp/+ mice. Collectively, our findings delineate potential alterations and correlations among the gut microbiota and microbial neurotransmitters in 16p11.2dp/+ mice, providing new insights into the pathogenesis of and treatment for 16p11.2 CNV-associated ASD.
Collapse
Affiliation(s)
- Zhang Fu
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiuyan Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Youheng Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xinliang Mao
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Hualin Liu
- Guangdong Perfect Life Health Science and Technology Research Institute Co., Ltd., Zhongshan, Guangdong, China
| | - Yanming Yang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jia Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhumei Chen
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Anesthesiology, The Seventh Affiliated Hospital of Sun Yat-Sen University (SYSU), Shenzhen, Guangdong, China
| | - Huiliang Li
- Division of Medicine, Wolfson Institute for Biomedical Research, Faculty of Medical Sciences, University College London, London, United Kingdom
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States
| | - Xinjun Mao
- Department of Anesthesiology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ningning Li
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- China-UK Institute for Frontier Science, Shenzhen, Guangdong, China
| | - Dilong Wang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Jiang
- Tomas Lindhal Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Berger SN, Baumberger B, Samaranayake S, Hersey M, Mena S, Bain I, Duncan W, Reed MC, Nijhout HF, Best J, Hashemi P. An In Vivo Definition of Brain Histamine Dynamics Reveals Critical Neuromodulatory Roles for This Elusive Messenger. Int J Mol Sci 2022; 23:14862. [PMID: 36499189 PMCID: PMC9738190 DOI: 10.3390/ijms232314862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/05/2022] Open
Abstract
Histamine is well known for mediating peripheral inflammation; however, this amine is also found in high concentrations in the brain where its roles are much less known. In vivo chemical dynamics are difficult to measure, thus fundamental aspects of histamine's neurochemistry remain undefined. In this work, we undertake the first in-depth characterization of real time in vivo histamine dynamics using fast electrochemical tools. We find that histamine release is sensitive to pharmacological manipulation at the level of synthesis, packaging, autoreceptors and metabolism. We find two breakthrough aspects of histamine modulation. First, differences in H3 receptor regulation between sexes show that histamine release in female mice is much more tightly regulated than in male mice under H3 or inflammatory drug challenge. We hypothesize that this finding may contribute to hormone-mediated neuroprotection mechanisms in female mice. Second, a high dose of a commonly available antihistamine, the H1 receptor inverse agonist diphenhydramine, rapidly decreases serotonin levels. This finding highlights the sheer significance of pharmaceuticals on neuromodulation. Our study opens the path to better understanding and treating histamine related disorders of the brain (such as neuroinflammation), emphasizing that sex and modulation (of serotonin) are critical factors to consider when studying/designing new histamine targeting therapeutics.
Collapse
Affiliation(s)
- Shane N. Berger
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | | | - Srimal Samaranayake
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
- Department of Physiology, Pharmacology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Sergio Mena
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Ian Bain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - William Duncan
- Department of Mathematics, Montana State University, Bozeman, MT 59717, USA
| | - Michael C. Reed
- Department of Mathematics, Duke University, Durham, NC 27710, USA
| | | | - Janet Best
- Department of Mathematics, Ohio State University, Columbus, OH 43210, USA
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
8
|
Salminen AV, Clemens S, García-Borreguero D, Ghorayeb I, Li Y, Manconi M, Ondo W, Rye D, Siegel JM, Silvani A, Winkelman JW, Allen RP, Ferré S. Consensus guidelines on the construct validity of rodent models of restless legs syndrome. Dis Model Mech 2022; 15:dmm049615. [PMID: 35946581 PMCID: PMC9393041 DOI: 10.1242/dmm.049615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/10/2022] [Indexed: 12/16/2022] Open
Abstract
Our understanding of the causes and natural course of restless legs syndrome (RLS) is incomplete. The lack of objective diagnostic biomarkers remains a challenge for clinical research and for the development of valid animal models. As a task force of preclinical and clinical scientists, we have previously defined face validity parameters for rodent models of RLS. In this article, we establish new guidelines for the construct validity of RLS rodent models. To do so, we first determined and agreed on the risk, and triggering factors and pathophysiological mechanisms that influence RLS expressivity. We then selected 20 items considered to have sufficient support in the literature, which we grouped by sex and genetic factors, iron-related mechanisms, electrophysiological mechanisms, dopaminergic mechanisms, exposure to medications active in the central nervous system, and others. These factors and biological mechanisms were then translated into rodent bioequivalents deemed to be most appropriate for a rodent model of RLS. We also identified parameters by which to assess and quantify these bioequivalents. Investigating these factors, both individually and in combination, will help to identify their specific roles in the expression of rodent RLS-like phenotypes, which should provide significant translational implications for the diagnosis and treatment of RLS.
Collapse
Affiliation(s)
- Aaro V. Salminen
- Institute of Neurogenomics, Helmholtz Zentrum München GmbH - German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | - Imad Ghorayeb
- Département de Neurophysiologie Clinique, Pôle Neurosciences Cliniques, CHU de Bordeaux, 33076 Bordeaux, France
- Université de Bordeaux, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, 33076 Bordeaux, France
- CNRS, Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, 33076 Bordeaux, France
| | - Yuqing Li
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mauro Manconi
- Sleep Medicine Unit, Regional Hospital of Lugano, Neurocenter of Southern Switzerland, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Department of Neurology, University Hospital Inselspital, 3010 Bern, Switzerland
| | - William Ondo
- Houston Methodist Hospital Neurological Institute, Weill Cornell Medical School, Houston, TX 77070, USA
| | - David Rye
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jerome M. Siegel
- Neuropsychiatric Institute and Brain Research Institute, University of California, Los Angeles, CA 90095, USA
- Neurobiology Research, Veterans Administration Greater Los Angeles Healthcare System, North Hills, CA 91343, USA
| | - Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences Alma Mater Studiorum, Università di Bologna, 48121 Ravenna Campus, Ravenna, Italy
| | - John W. Winkelman
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richard P. Allen
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | | |
Collapse
|
9
|
Dela Peña IC, Figueroa JD, Shi WX. Hypothesis: Amelioration of obesity-induced cognitive dysfunction via a lorcaserin-betahistine combination treatment. Pharmacol Res Perspect 2022; 10:e00947. [PMID: 35599337 PMCID: PMC9124816 DOI: 10.1002/prp2.947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/11/2022] [Indexed: 11/12/2022] Open
Abstract
The prolonged exposure to obesogenic diets disrupts the mesocortical dopaminergic input to the prefrontal cortex (PFC). This leads to suboptimal dopamine levels in this brain region, which affects cognition and control of food intake. Treatments that restore mesocortical dopaminergic neurotransmission may improve obesity‐associated cognitive dysfunction and modulate food intake to induce weight loss. Given the complexity and multifactorial nature of obesity, combination treatments would likely achieve sizeable and sustained body weight loss and improve obesity‐linked outcomes, such as cognitive dysfunction. Given this background, we hypothesize that concomitant activation of serotonin 5‐HT2C and histamine H1 receptors, coupled with antagonism of histamine H3 receptors, synergistically modulates mesocortical dopamine neurotransmission and ameliorates obesity‐induced cognitive dysfunction. We propose to test the hypothesis in a diet‐induced obesity (DIO) rat model by treating animals with the 5‐HT2C agonist lorcaserin and the H1 agonist and H3 antagonist betahistine. Consistent with our hypothesis, both lorcaserin and betahistine have been shown to reduce body weight in humans with obesity and animals. Both drugs have been demonstrated to improve cognitive functions by influencing dopaminergic signaling in the PFC. The proposed combination treatment addresses the paucity of studies on obesity treatments that improve cognitive function. This research may also help identify a potential targetable mechanism connecting obesity and neurocognitive outcomes.
Collapse
Affiliation(s)
- Ike C Dela Peña
- Departments of Pharmaceutical and Administrative Sciences, Loma Linda University Schools of Pharmacy, Loma Linda, California, USA
| | - Johnny D Figueroa
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Physiology Division, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Wei-Xing Shi
- Departments of Pharmaceutical and Administrative Sciences, Loma Linda University Schools of Pharmacy, Loma Linda, California, USA.,Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
10
|
Beheshti S, Wasil Wesal M. Anticonvulsant activity of the histamine H3 receptor inverse agonist pitolisant in an electrical kindling model of epilepsy. Neurosci Lett 2022; 782:136685. [DOI: 10.1016/j.neulet.2022.136685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
|
11
|
Arrigoni E, Fuller PM. The Role of the Central Histaminergic System in Behavioral State Control. Curr Top Behav Neurosci 2022; 59:447-468. [PMID: 34595740 DOI: 10.1007/7854_2021_263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Histamine is a small monoamine signaling molecule that plays a role in many peripheral and central physiological processes, including the regulation of wakefulness. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and histamine neurons are thought to promote wakefulness and vigilance maintenance - under certain environmental and/or behavioral contexts - through their diffuse innervation of the cortex and other wake-promoting brain circuits. Histamine neurons also contain a number of other putative neurotransmitters, although the functional role of these co-transmitters remains incompletely understood. Within the brain histamine operates through three receptor subtypes that are located on pre- and post-synaptic membranes. Some histamine receptors exhibit constitutive activity, and hence exist in an activated state even in the absence of histamine. Newer medications used to reduce sleepiness in narcolepsy patients in fact enhance histamine signaling by blunting the constitutive activity of these histamine receptors. In this chapter, we provide an overview of the central histamine system with an emphasis on its role in behavioral state regulation and how drugs targeting histamine receptors are used clinically to treat a wide range of sleep-wake disorders.
Collapse
Affiliation(s)
- Elda Arrigoni
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Patrick M Fuller
- Department of Neurological Surgery, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
12
|
Pandey M, Bhati A, Priya K, Sharma KK, Singhal B. Precision Postbiotics and Mental Health: the Management of Post-COVID-19 Complications. Probiotics Antimicrob Proteins 2021; 14:426-448. [PMID: 34806151 PMCID: PMC8606251 DOI: 10.1007/s12602-021-09875-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/14/2023]
Abstract
The health catastrophe originated by COVID-19 pandemic construed profound impact on a global scale. However, a plethora of research studies corroborated convincing evidence conferring severity of infection of SARS-CoV-2 with the aberrant gut microbiome that strongly speculated its importance for development of novel therapeutic modalities. The intense exploration of probiotics has been envisaged to promote the healthy growth of the host, and restore intestinal microecological balance through various metabolic and physiological processes. The demystifying effect of probiotics cannot be defied, but there exists a strong skepticism related to their safety and efficacy. Therefore, molecular signature of probiotics termed as "postbiotics" are of paramount importance and there is continuous surge of utilizing postbiotics for enhancing health benefits, but little is explicit about their antiviral effects. Therefore, it is worth considering their prospective role in post-COVID regime that pave the way for exploring the pastoral vistas of postbiotics. Based on previous research investigations, the present article advocates prospective role of postbiotics in alleviating the health burden of viral infections, especially SARS-CoV-2. The article also posits current challenges and proposes a futuristic model describing the concept of "precision postbiotics" for effective therapeutic and preventive interventions that can be used for management of this deadly disease.
Collapse
Affiliation(s)
- Muskan Pandey
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Archana Bhati
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Kumari Priya
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - K K Sharma
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Barkha Singhal
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India.
| |
Collapse
|
13
|
Effect of Central Administration of Brain-Derived Neurotrophic Factor (BDNF) on Behavior and Brain Monoamine Metabolism in New Recombinant Mouse Lines Differing by 5-HT 1A Receptor Functionality. Int J Mol Sci 2021; 22:ijms222111987. [PMID: 34769417 PMCID: PMC8584822 DOI: 10.3390/ijms222111987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 11/29/2022] Open
Abstract
Experiments were carried out on recombinant B6.CBA-D13Mit76C (B6-M76C) and B6.CBA-D13Mit76B (B6-M76B) mouse lines created by transferring a 102.73–118.83 Mbp fragment of chromosome 13, containing the 5-HT1A receptor gene, from CBA or C57BL/6 strains to a C57BL/6 genetic background, correspondingly. We have recently shown different levels of 5-HT1A receptor functionality in these mouse lines. The administration of BDNF (300 ng/mouse, i.c.v.) increased the levels of exploratory activity and intermale aggression only in B6-M76B mice, without affecting depressive-like behavior in both lines. In B6-M76B mice the behavioral alterations were accompanied by a decrease in the 5-HT2A receptor functional activity and the augmentation of levels of serotonin and its main metabolite, 5-HIAA (5-hydroxyindoleacetic acid), in the midbrain. Moreover, the levels of dopamine and its main metabolites, HVA (homovanillic acid) and DOPAC (3,4-dihydroxyphenylacetic acid), were also elevated in the striatum of B6-M76B mice after BDNF treatment. In B6-M76C mice, central BDNF administration led only to a reduction in the functional activity of the 5-HT1A receptor and a rise in DOPAC levels in the midbrain. The obtained data suggest the importance of the 102.73–118.83 Mbp fragment of mouse chromosome 13, which contains the 5-HT1A receptor gene, for BDNF-induced alterations in behavior and the brain monoamine system.
Collapse
|
14
|
Yang L, Wang Y, Chen Z. Central histaminergic signalling, neural excitability and epilepsy. Br J Pharmacol 2021; 179:3-22. [PMID: 34599508 DOI: 10.1111/bph.15692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/31/2022] Open
Abstract
Epilepsy is a common neurological disorder characterized by repeated and spontaneous epileptic seizures and is not well controlled by current medication. Traditional theory suggests that epilepsy results from an imbalance of excitatory glutamate neurons and inhibitory GABAergic neurons. However, new evidence from clinical and preclinical research suggests that histamine in the CNS plays an important role in the modulation of neural excitability and in the pathogenesis of epilepsy. Many histamine receptor ligands have achieved curative effects in animal epilepsy models, among which the histamine H3 receptor antagonist pitolisant has shown anti-epileptic effects in clinical trials. Recent studies, therefore, have focused on the potential action of histamine receptors to control and treat epilepsy. In this review, we summarize the findings from animal and clinical epilepsy research on the role of brain histamine and its receptors. We also identify current gaps in the research and suggest where further studies are most needed.
Collapse
Affiliation(s)
- Lin Yang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
15
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
16
|
Hersey M, Hashemi P, Reagan LP. Integrating the monoamine and cytokine hypotheses of depression: Is histamine the missing link? Eur J Neurosci 2021; 55:2895-2911. [PMID: 34265868 DOI: 10.1111/ejn.15392] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022]
Abstract
Psychiatric diseases, like depression, largely affect the central nervous system (CNS). While the underlying neuropathology of depressive illness remains to be elucidated, several hypotheses have been proposed as molecular underpinnings for major depressive disorder, including the monoamine hypothesis and the cytokine hypothesis. The monoamine hypothesis has been largely supported by the pharmaceuticals that target monoamine neurotransmitters as a treatment for depression. However, these antidepressants have come under scrutiny due to their limited clinical efficacy, side effects, and delayed onset of action. The more recent, cytokine hypothesis of depression is supported by the ability of immune-active agents to induce "sickness behaviour" akin to that seen with depression. However, treatments that more selectively target inflammation have yielded inconsistent antidepressive results. As such, neither of these hypotheses can fully explain depressive illness pathology, implying that the underlying neuropathological mechanisms may encompass aspects of both theories. The goal of the current review is to integrate these two well-studied hypotheses and to propose a role for histamine as a potential unifying factor that links monoamines to cytokines. Additionally, we will focus on stress-induced depression, to provide an updated perspective of depressive illness research and thereby identify new potential targets for the treatment of major depressive disorder.
Collapse
Affiliation(s)
- Melinda Hersey
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina, USA
| | - Parastoo Hashemi
- Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina, USA.,Department of Bioengineering, Imperial College, London, UK
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA.,WJB Dorn Veterans Affairs Medical Center, Columbia, South Carolina, USA
| |
Collapse
|
17
|
Grinchii D, Dremencov E. Mechanism of Action of Atypical Antipsychotic Drugs in Mood Disorders. Int J Mol Sci 2020; 21:ijms21249532. [PMID: 33333774 PMCID: PMC7765178 DOI: 10.3390/ijms21249532] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 01/07/2023] Open
Abstract
Atypical antipsychotic drugs were introduced in the early 1990s. Unlike typical antipsychotics, which are effective only against positive symptoms of schizophrenia, atypical antipsychotics are effective against negative and cognitive symptoms as well. Furthermore, they are effective not only in psychotic but also in affective disorders, on their own or as adjuncts to antidepressant drugs. This review presents the neural mechanisms of currently existing atypical antipsychotics and putative antipsychotics currently being investigated in preclinical and clinical studies and how these relate to their effectiveness in mood disorders such as depression, anxiety, and post-traumatic stress disorder (PTSD). Typical antipsychotics act almost exclusively on the dopamine system. Atypical drugs, however, modulate serotonin (5-HT), norepinephrine, and/or histamine neurotransmission as well. This multimodal mechanism of action putatively underlies the beneficial effect of atypical antipsychotics in mood and anxiety disorders. Interestingly, novel experimental drugs having dual antipsychotic and antidepressant therapeutic potential, such as histamine, adenosine, and trace amine-associated receptors (TAAR) ligand, are also characterized by a multimodal stimulatory effect on central 5-HT, norepinephrine, and/or histamine transmission. The multimodal stimulatory effect on central monoamine neurotransmission may be thus primarily responsible for the combined antidepressant and antipsychotic therapeutic potential of certain central nervous system (CNS) drugs.
Collapse
|
18
|
Labouesse MA, Cola RB, Patriarchi T. GPCR-Based Dopamine Sensors-A Detailed Guide to Inform Sensor Choice for In vivo Imaging. Int J Mol Sci 2020; 21:E8048. [PMID: 33126757 PMCID: PMC7672611 DOI: 10.3390/ijms21218048] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding how dopamine (DA) encodes behavior depends on technologies that can reliably monitor DA release in freely-behaving animals. Recently, red and green genetically encoded sensors for DA (dLight, GRAB-DA) were developed and now provide the ability to track release dynamics at a subsecond resolution, with submicromolar affinity and high molecular specificity. Combined with rapid developments in in vivo imaging, these sensors have the potential to transform the field of DA sensing and DA-based drug discovery. When implementing these tools in the laboratory, it is important to consider there is not a 'one-size-fits-all' sensor. Sensor properties, most importantly their affinity and dynamic range, must be carefully chosen to match local DA levels. Molecular specificity, sensor kinetics, spectral properties, brightness, sensor scaffold and pharmacology can further influence sensor choice depending on the experimental question. In this review, we use DA as an example; we briefly summarize old and new techniques to monitor DA release, including DA biosensors. We then outline a map of DA heterogeneity across the brain and provide a guide for optimal sensor choice and implementation based on local DA levels and other experimental parameters. Altogether this review should act as a tool to guide DA sensor choice for end-users.
Collapse
Affiliation(s)
- Marie A. Labouesse
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA;
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Reto B. Cola
- Anatomy and Program in Neuroscience, University of Fribourg, 1700 Fribourg, Switzerland;
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
19
|
Zhang X, Baer AG, Price JM, Jones PC, Garcia BJ, Romero J, Cliff AM, Mi W, Brown JB, Jacobson DA, Lydic R, Baghdoyan HA. Neurotransmitter networks in mouse prefrontal cortex are reconfigured by isoflurane anesthesia. J Neurophysiol 2020; 123:2285-2296. [PMID: 32347157 PMCID: PMC7311717 DOI: 10.1152/jn.00092.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/13/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
This study quantified eight small-molecule neurotransmitters collected simultaneously from prefrontal cortex of C57BL/6J mice (n = 23) during wakefulness and during isoflurane anesthesia (1.3%). Using isoflurane anesthesia as an independent variable enabled evaluation of the hypothesis that isoflurane anesthesia differentially alters concentrations of multiple neurotransmitters and their interactions. Machine learning was applied to reveal higher order interactions among neurotransmitters. Using a between-subjects design, microdialysis was performed during wakefulness and during anesthesia. Concentrations (nM) of acetylcholine, adenosine, dopamine, GABA, glutamate, histamine, norepinephrine, and serotonin in the dialysis samples are reported (means ± SD). Relative to wakefulness, acetylcholine concentration was lower during isoflurane anesthesia (1.254 ± 1.118 vs. 0.401 ± 0.134, P = 0.009), and concentrations of adenosine (29.456 ± 29.756 vs. 101.321 ± 38.603, P < 0.001), dopamine (0.0578 ± 0.0384 vs. 0.113 ± 0.084, P = 0.036), and norepinephrine (0.126 ± 0.080 vs. 0.219 ± 0.066, P = 0.010) were higher during anesthesia. Isoflurane reconfigured neurotransmitter interactions in prefrontal cortex, and the state of isoflurane anesthesia was reliably predicted by prefrontal cortex concentrations of adenosine, norepinephrine, and acetylcholine. A novel finding to emerge from machine learning analyses is that neurotransmitter concentration profiles in mouse prefrontal cortex undergo functional reconfiguration during isoflurane anesthesia. Adenosine, norepinephrine, and acetylcholine showed high feature importance, supporting the interpretation that interactions among these three transmitters may play a key role in modulating levels of cortical and behavioral arousal.NEW & NOTEWORTHY This study discovered that interactions between neurotransmitters in mouse prefrontal cortex were altered during isoflurane anesthesia relative to wakefulness. Machine learning further demonstrated that, relative to wakefulness, higher order interactions among neurotransmitters were disrupted during isoflurane administration. These findings extend to the neurochemical domain the concept that anesthetic-induced loss of wakefulness results from a disruption of neural network connectivity.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee
- Department of Psychology, University of Tennessee, Knoxville, Tennessee
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - Aaron G Baer
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Joshua M Price
- Office of Information Technology, University of Tennessee, Knoxville, Tennessee
| | - Piet C Jones
- Oak Ridge National Laboratory, Oak Ridge, Tennessee
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee
| | | | - Jonathon Romero
- Oak Ridge National Laboratory, Oak Ridge, Tennessee
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee
| | - Ashley M Cliff
- Oak Ridge National Laboratory, Oak Ridge, Tennessee
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, Tennessee
| | - Weidong Mi
- Anesthesia and Operation Center, Chinese PLA General Hospital, Beijing, China
| | - James B Brown
- Molecular Ecosystems Biology Department, Lawrence Berkeley National Laboratory, Berkeley, California
| | | | - Ralph Lydic
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee
- Department of Psychology, University of Tennessee, Knoxville, Tennessee
- Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Helen A Baghdoyan
- Department of Anesthesiology, University of Tennessee Medical Center, Knoxville, Tennessee
- Department of Psychology, University of Tennessee, Knoxville, Tennessee
- Oak Ridge National Laboratory, Oak Ridge, Tennessee
| |
Collapse
|
20
|
Brunet A, Stuart-Lopez G, Burg T, Scekic-Zahirovic J, Rouaux C. Cortical Circuit Dysfunction as a Potential Driver of Amyotrophic Lateral Sclerosis. Front Neurosci 2020; 14:363. [PMID: 32410944 PMCID: PMC7201269 DOI: 10.3389/fnins.2020.00363] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that affects selected cortical and spinal neuronal populations, leading to progressive paralysis and death. A growing body of evidences suggests that the disease may originate in the cerebral cortex and propagate in a corticofugal manner. In particular, transcranial magnetic stimulation studies revealed that ALS patients present with early cortical hyperexcitability arising from a combination of increased excitability and decreased inhibition. Here, we discuss the possibility that initial cortical circuit dysfunction might act as the main driver of ALS onset and progression, and review recent functional, imaging and transcriptomic studies conducted on ALS patients, along with electrophysiological, pathological and transcriptomic studies on animal and cellular models of the disease, in order to evaluate the potential cellular and molecular origins of cortical hyperexcitability in ALS.
Collapse
Affiliation(s)
| | | | | | | | - Caroline Rouaux
- INSERM UMR_S 1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
21
|
Midzyanovskaya IS, Shatskova AB, MacDonald E, Luijtelaar GV, Tuomisto L. Brain Aminergic Deficiency in Absence Epileptic Rats: Dependency on Seizure Severity and Their Functional Coupling at Rest. ACTA ACUST UNITED AC 2020. [DOI: 10.4236/jbbs.2020.101003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Scammell TE, Jackson AC, Franks NP, Wisden W, Dauvilliers Y. Histamine: neural circuits and new medications. Sleep 2019; 42:5099478. [PMID: 30239935 PMCID: PMC6335869 DOI: 10.1093/sleep/zsy183] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Indexed: 12/12/2022] Open
Abstract
Histamine was first identified in the brain about 50 years ago, but only in the last few years have researchers gained an understanding of how it regulates sleep/wake behavior. We provide a translational overview of the histamine system, from basic research to new clinical trials demonstrating the usefulness of drugs that enhance histamine signaling. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and like many of the arousal systems, histamine neurons diffusely innervate the cortex, thalamus, and other wake-promoting brain regions. Histamine has generally excitatory effects on target neurons, but paradoxically, histamine neurons may also release the inhibitory neurotransmitter GABA. New research demonstrates that activity in histamine neurons is essential for normal wakefulness, especially at specific circadian phases, and reducing activity in these neurons can produce sedation. The number of histamine neurons is increased in narcolepsy, but whether this affects brain levels of histamine is controversial. Of clinical importance, new compounds are becoming available that enhance histamine signaling, and clinical trials show that these medications reduce sleepiness and cataplexy in narcolepsy.
Collapse
Affiliation(s)
- Thomas E Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Alexander C Jackson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT
| | - Nicholas P Franks
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, UK
| | - William Wisden
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, UK
| | - Yves Dauvilliers
- Centre National de Référence Narcolepsie Hypersomnies, Unité des Troubles du Sommeil, Service de Neurologie, Hôpital Gui-de-Chauliac, Université Montpellier, INSERM, Montpellier, France
| |
Collapse
|
23
|
TAK-925, an orexin 2 receptor-selective agonist, shows robust wake-promoting effects in mice. Pharmacol Biochem Behav 2019; 187:172794. [DOI: 10.1016/j.pbb.2019.172794] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 11/21/2022]
|
24
|
Koprdova R, Csatlosova K, Durisova B, Bogi E, Majekova M, Dremencov E, Mach M. Electrophysiology and Behavioral Assessment of the New Molecule SMe1EC2M3 as a Representative of the Future Class of Triple Reuptake Inhibitors. Molecules 2019; 24:molecules24234218. [PMID: 31757051 PMCID: PMC6930491 DOI: 10.3390/molecules24234218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 11/16/2022] Open
Abstract
SMe1EC2M3 is a pyridoindole derivative related to the neuroleptic drug carbidine. Based on the structural similarities of SMe1EC2M3 and known serotonin (5-HT), norepinephrine, and dopamine reuptake inhibitors, we hypothesized that this compound may also have triple reuptake inhibition efficacy and an antidepressant-like effect. PreADMET and Dragon software was used for in silico prediction of pharmacokinetics and pharmacodynamics of SMe1EC2M3. Forced swim test was used to evaluate its antidepressant-like effects. Extracellular in vivo electrophysiology was used to assess 5-HT, norepinephrine, and dopamine reuptake inhibition efficacy of SMe1EC2M3. PreADMET predicted reasonable intestinal absorption, plasma protein binding, and blood-brain permeability for SMe1EC2M3. Dragon forecasted its efficiency as an antidepressant. Using behavioral measurements, it was found that SMe1EC2M3 decreased immobility time and increase swimming time during the forced swim test (FST). Electrophysiological investigations showed that SMe1EC2M3 dose-dependently suppressed the excitability of 5-HT neurons of the dorsal raphe nucleus (DRN), norepinephrine neurons of the locus coeruleus (LC), and dopamine neurons of the ventral tegmental area (VTA). The SMe1EC2M3-induced suppression of 5-HT, norepinephrine, and dopamine neurons was reversed by the antagonists of serotonin-1A (5-HT1A; WAY100135), α-2 adrenergic (α2, yohimbine), and dopamine-2 receptors (D2, haloperidol), respectively. We conclude that SMe1EC2M3 is prospective triple 5-HT, norepinephrine, and dopamine reuptake inhibitor with antidepressant-like properties, however future studies should be performed to complete the pharmacological profiling of this compound.
Collapse
Affiliation(s)
- Romana Koprdova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia; (R.K.)
| | - Kristina Csatlosova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia; (R.K.)
| | - Barbora Durisova
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| | - Eszter Bogi
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia; (R.K.)
| | - Magdalena Majekova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia; (R.K.)
| | - Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Center of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovakia
| | - Mojmir Mach
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, Dúbravská cesta 9, 841 04 Bratislava, Slovakia; (R.K.)
- Correspondence: ; Tel.: +421-2-3229-5718
| |
Collapse
|
25
|
Puthongkham P, Lee ST, Venton BJ. Mechanism of Histamine Oxidation and Electropolymerization at Carbon Electrodes. Anal Chem 2019; 91:8366-8373. [PMID: 31194511 DOI: 10.1021/acs.analchem.9b01178] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Histamine plays an important role in neuromodulation and the biological immune response. Although many electrochemical methods have been developed for histamine detection, the mechanism of its redox reaction has not been directly investigated. Here, we studied the mechanism of histamine oxidation at carbon electrodes and used that mechanistic information to design better fast-scan cyclic voltammetry (FSCV) methods for histamine. Using amperometry, cyclic voltammetry (CV), and X-ray photoelectron spectroscopy (XPS), we demonstrate that histamine oxidation requires a potential of at least +1.1 V vs Ag/AgCl. We propose that histamine undergoes one-electron oxidation on an imidazole nitrogen that produces a radical. The radical species dimerize and continue to undergo oxidation, leading to electropolymerization, which fouls the electrode. CV shows a peak at 1.3 V that is pH dependent, consistent with a one-proton, one-electron oxidation reaction. This mechanism is confirmed using 1- and 3-methylhistamine, which do not electropolymerize, compared to Nα-methylhistamine, which does. XPS also revealed a nitrogen-containing product adsorbed on the electrode surface after histamine oxidation. For FSCV detection of histamine at carbon-fiber microelectrodes, histamine oxidation was adsorption-controlled, and the anodic peak was observed at +1.2 V on the backward scan because of the rapid scan rate. However, the oxidation fouled the electrode and convoluted the FSCV temporal response; therefore, we implemented Nafion coating to alleviate the electrode fouling and preserve the time response of FSCV. Knowing the mechanism of histamine oxidation will facilitate design of better electrochemical methods for real-time monitoring of histamine.
Collapse
Affiliation(s)
- Pumidech Puthongkham
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| | - Scott T Lee
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| | - B Jill Venton
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904 , United States
| |
Collapse
|
26
|
Hassan A, Robinson M, Willerth SM. Modeling the Effects of Yoga on the Progression of Alzheimer's Disease in a Dish. Cells Tissues Organs 2019; 206:263-271. [PMID: 31121578 DOI: 10.1159/000499503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 03/11/2019] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) accounts for 80% of all dementia cases, making it the most common form of dementia. Aging serves as the main risk factor for AD, but early onset AD can also occur in individuals younger than 65 years. AD results from progressive neurodegeneration leading to dysfunctional synaptic transmission in the brain. The cascade hypothesis of AD states that amyloid precursor protein (APP) metabolism becomes impaired either by mutation or an interleukin-mediated stress response to injury, resulting in the splicing of harmful oligomeric forms of amyloid beta (Aβ). These oligomers disrupt extracellular receptor binding, intracellular function, and cellular membrane integrity. Yoga and meditative practices slow the progression of the cognitive decline associated with AD. However, the biological mechanisms underlying this therapeutic effect remain elusive. Here, we investigated the ability of neurotransmitters released during yoga and meditative practices to rescue neurons from synaptic dysfunction in an in vitro Alzheimer's model created by culturing basal forebrain cholinergic neurons with physiologically relevant levels of the I-42 isoform of oligomeric Aβ (OΑβI-42). We found that the neurotransmitters dopamine and histamine produce a cooperative action with serotonin to reverse the loss of choline acetyltransferase (CHaT) by OΑβI-42. The loss of ChaT, the enzyme responsible for processing the cholinergic neurotransmitter acetylcholine, contributes to the synaptic dysfunction experienced during AD. These neurotransmitters inhibit nitric oxide synthesis caused by OΑβI-42, preventing oxidative and nitrosative stress. Serotonin activates an alternate cleavage of APP to produce a fragment with known neurotrophic effects, giving it the unique ability to inhibit the OΑβI-42 production cycle. We hypothesize here that these concerted actions lead to the protection of cholinergic synaptic transmission in AD.
Collapse
Affiliation(s)
- Adithy Hassan
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Meghan Robinson
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Stephanie M Willerth
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada, .,Biomedical Engineering Program, University of Victoria, Victoria, British Columbia, Canada, .,Department of Mechanical Engineering, Faculty of Engineering, University of Victoria, Victoria, British Columbia, Canada, .,Centre for Biomedical Research, Faculty of Engineering, University of Victoria, Victoria, British Columbia, Canada, .,International Collaboration for Repair Discovery, University of British Columbia, Vancouver, British Columbia, Canada,
| |
Collapse
|
27
|
Yu X, Ma Y, Harding EC, Yustos R, Vyssotski AL, Franks NP, Wisden W. Genetic lesioning of histamine neurons increases sleep-wake fragmentation and reveals their contribution to modafinil-induced wakefulness. Sleep 2019; 42:zsz031. [PMID: 30722053 PMCID: PMC6519916 DOI: 10.1093/sleep/zsz031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/22/2019] [Accepted: 01/29/2019] [Indexed: 11/12/2022] Open
Abstract
Acute chemogenetic inhibition of histamine (HA) neurons in adult mice induced nonrapid eye movement (NREM) sleep with an increased delta power. By contrast, selective genetic lesioning of HA neurons with caspase in adult mice exhibited a normal sleep-wake cycle overall, except at the diurnal start of the lights-off period, when they remained sleepier. The amount of time spent in NREM sleep and in the wake state in mice with lesioned HA neurons was unchanged over 24 hr, but the sleep-wake cycle was more fragmented. Both the delayed increase in wakefulness at the start of the night and the sleep-wake fragmentation are similar phenotypes to histidine decarboxylase knockout mice, which cannot synthesize HA. Chronic loss of HA neurons did not affect sleep homeostasis after sleep deprivation. However, the chronic loss of HA neurons or chemogenetic inhibition of HA neurons did notably reduce the ability of the wake-promoting compound modafinil to sustain wakefulness. Thus, part of modafinil's wake-promoting actions arise through the HA system.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Life Sciences, Imperial College London, UK
| | - Ying Ma
- Department of Life Sciences, Imperial College London, UK
| | | | - Raquel Yustos
- Department of Life Sciences, Imperial College London, UK
| | - Alexei L Vyssotski
- Institute of Neuroinformatics, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, UK
- UK Dementia Research Institute at Imperial College London, UK
| | - William Wisden
- Department of Life Sciences, Imperial College London, UK
- UK Dementia Research Institute at Imperial College London, UK
| |
Collapse
|
28
|
Li Y, He R, Niu Y, Li F. Paper-Based Electrochemical Biosensors for Point-of-Care Testing of Neurotransmitters. JOURNAL OF ANALYSIS AND TESTING 2019. [DOI: 10.1007/s41664-019-00085-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Patnaik R, Sharma A, Skaper SD, Muresanu DF, Lafuente JV, Castellani RJ, Nozari A, Sharma HS. Histamine H3 Inverse Agonist BF 2649 or Antagonist with Partial H4 Agonist Activity Clobenpropit Reduces Amyloid Beta Peptide-Induced Brain Pathology in Alzheimer's Disease. Mol Neurobiol 2019; 55:312-321. [PMID: 28861757 DOI: 10.1007/s12035-017-0743-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Alzheimer's disease (AD) is one of the leading causes for disability and death affecting millions of people worldwide. Thus, novel therapeutic strategies are needed to reduce brain pathology associated with AD. In view of increasing awareness regarding involvement of histaminergic pathways in AD, we explored the role of one H3 receptor inverse agonist BF 2649 and one selective H3 receptor antagonist with partial H4 agonist activity in amyloid beta peptide (AβP) infusion-induced brain pathology in a rat model. AD-like pathology was produced by administering AβP (1-40) intracerebroventricular (i.c.v.) in the left lateral ventricle (250 ng/10 μl, once daily) for 4 weeks. Control rats received saline. In separate group of rats, either BF 2649 (1 mg/kg, i.p.) or clobenpropit (1 mg/kg, i.p.) was administered once daily for 1 week after 3 weeks of AβP administration. After 30 days, blood-brain barrier (BBB) breakdown, edema formation, neuronal, glial injuries, and AβP deposits were examined in the brain. A significant reduction in AβP deposits along with marked reduction in neuronal or glial reactions was seen in the drug-treated group. The BBB breakdown to Evans blue albumin and radioiodine in the cortex, hippocampus, hypothalamus, and cerebellum was also significantly reduced in these drug-treated groups. Clobenpropit showed superior effects than the BF2649 in reducing brain pathology in AD. Taken together, our observations are the first to show that blockade of H3 and stimulation of H4 receptors are beneficial for the treatment of AD pathology, not reported earlier.
Collapse
Affiliation(s)
- Ranjana Patnaik
- School of Biomedical Engineering, Department of Biomaterials, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Uppsala University Hospital, Uppsala University, SE-75185, Uppsala, Sweden
| | - Aruna Sharma
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Uppsala University Hospital, Uppsala University, SE-75185, Uppsala, Sweden
- International Experimental Central Nervous System Injury and Repair (IECNSIR), University Hospital, Uppsala University, Frödingsgatan 12, Bldg. 28, SE-75421, Uppsala, Sweden
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Dafin F Muresanu
- "RoNeuro" Institute for Neurological Research and Diagnostic, 37 Mircea Eliade Street, 400364, Cluj-Napoca, Romania
- Department of Clinical Neurosciences, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
- Nanoneurosurgery Group, BioCruces Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
- Faculty of Health Science, Universidad Autónoma de Chile, Santiago de Chile, Chile
| | | | - Ala Nozari
- Anesthesiology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Hari S Sharma
- Department of Surgical Sciences, Anesthesiology and Intensive Care Medicine, Uppsala University Hospital, Uppsala University, SE-75185, Uppsala, Sweden.
- International Experimental Central Nervous System Injury and Repair (IECNSIR), University Hospital, Uppsala University, Frödingsgatan 12, Bldg. 28, SE-75421, Uppsala, Sweden.
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.
| |
Collapse
|
30
|
van den Brink WJ, Hartman R, van den Berg D, Flik G, Gonzalez‐Amoros B, Koopman N, Elassais‐Schaap J, van der Graaf PH, Hankemeier T, de Lange EC. Blood-Based Biomarkers of Quinpirole Pharmacology: Cluster-Based PK/PD and Metabolomics to Unravel the Underlying Dynamics in Rat Plasma and Brain. CPT Pharmacometrics Syst Pharmacol 2019; 8:107-117. [PMID: 30680960 PMCID: PMC6389346 DOI: 10.1002/psp4.12370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A key challenge in the development of central nervous system drugs is the availability of drug target specific blood-based biomarkers. As a new approach, we applied cluster-based pharmacokinetic/pharmacodynamic (PK/PD) analysis in brain extracellular fluid (brainECF ) and plasma simultaneously after 0, 0.17, and 0.86 mg/kg of the dopamine D2/3 agonist quinpirole (QP) in rats. We measured 76 biogenic amines in plasma and brainECF after single and 8-day administration, to be analyzed by cluster-based PK/PD analysis. Multiple concentration-effect relations were observed with potencies ranging from 0.001-383 nM. Many biomarker responses seem to distribute over the blood-brain barrier (BBB). Effects were observed for dopamine and glutamate signaling in brainECF , and branched-chain amino acid metabolism and immune signaling in plasma. Altogether, we showed for the first time how cluster-based PK/PD could describe a systems-response across plasma and brain, thereby identifying potential blood-based biomarkers. This concept is envisioned to provide an important connection between drug discovery and early drug development.
Collapse
Affiliation(s)
- Willem J. van den Brink
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Robin Hartman
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Dirk‐Jan van den Berg
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | - Belén Gonzalez‐Amoros
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Nanda Koopman
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Jeroen Elassais‐Schaap
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Piet Hein van der Graaf
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
- Certara QSPCanterbury Innovation HouseCanterburyUK
| | - Thomas Hankemeier
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Elizabeth C.M. de Lange
- Division of Systems Biomedicine and PharmacologyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
31
|
Verma L, Agrawal D, Jain NS. Enhanced central histaminergic transmission attenuates compulsive-like behavior in mice. Neuropharmacology 2018; 138:106-117. [DOI: 10.1016/j.neuropharm.2018.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022]
|
32
|
Olszewska B, Stasiak A, McNaught Flores D, Fogel WA, Leurs R, Walczyński K. 4-Hydroxypiperidines and Their Flexible 3-(Amino)propyloxy Analogues as Non-Imidazole Histamine H₃ Receptor Antagonist: Further Structure⁻Activity Relationship Exploration and In Vitro and In Vivo Pharmacological Evaluation. Int J Mol Sci 2018; 19:ijms19041243. [PMID: 29671795 PMCID: PMC5979327 DOI: 10.3390/ijms19041243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/05/2018] [Accepted: 04/17/2018] [Indexed: 11/30/2022] Open
Abstract
Presynaptic histamine H3 receptors (H3R) act as auto- or heteroreceptors controlling, respectively, the release of histamine and of other neurotransmitters in the central nervous system (CNS). The extracellular levels of several neurotransmitters are enhanced by H3R antagonists, and there is a great interest for potent, brain-penetrating H3 receptor antagonists/inverse agonists to compensate for the neurotransmitter deficits present in various neurological disorders. We have shown that 1-[(benzylfuran-2-yl)methyl]piperidinyl-4-oxyl- and benzyl- derivatives of N-propylpentan-1-amines exhibit high in vitro potencies toward the guinea pig H3 receptor (jejunum), with pA2 = 8.47 and 7.79, respectively (the reference compound used was thioperamide with pA2 = 8.67). Furthermore, following the replacement of 4-hydroxypiperidine with a 3-(methylamino)propyloxy chain, the pA2 value for the first group decreased, whereas it increased for the second group. Here, we present data on the impact of elongating the aliphatic chain between the nitrogen of 4-hydroxypiperidine or 3-(methylamino)propan-1-ol and the lipophilic residue. Additionally, the most active compound in this series of non-imidazole H3 receptor antagonists/inverse agonists, i.e., ADS-003, was evaluated for its affinity to the recombinant rat and human histamine H3 receptors transiently expressed in HEK-293T cells. It was shown that ADS-003, given parenterally for 5 days, reduced the food intake of rats, as well as changed histamine and noradrenaline concentrations in the rats’ brain in a manner and degree similar to the reference H3 antagonist Ciproxifan.
Collapse
Affiliation(s)
- Beata Olszewska
- Department of Synthesis and Technology of Drugs, Medical University of Lodz, Muszyńskiego Street 1, 90-145 Łódź, Poland.
| | - Anna Stasiak
- Department of Hormone Biochemistry, Medical University of Lodz, Żeligowskiego Street 7/9, 90-752 Łódź, Poland.
| | - Daniel McNaught Flores
- Amsterdam Institute of Molecules, Medicines & Systems, Division of Medicinal Chemistry, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| | - Wiesława Agnieszka Fogel
- Department of Hormone Biochemistry, Medical University of Lodz, Żeligowskiego Street 7/9, 90-752 Łódź, Poland.
| | - Rob Leurs
- Amsterdam Institute of Molecules, Medicines & Systems, Division of Medicinal Chemistry, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands.
| | - Krzysztof Walczyński
- Department of Synthesis and Technology of Drugs, Medical University of Lodz, Muszyńskiego Street 1, 90-145 Łódź, Poland.
| |
Collapse
|
33
|
Puttonen HAJ, Sundvik M, Semenova S, Shirai Y, Chen YC, Panula P. Knockout of histamine receptor H3 alters adaptation to sudden darkness and monoamine levels in the zebrafish. Acta Physiol (Oxf) 2018; 222. [PMID: 29044927 DOI: 10.1111/apha.12981] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 12/13/2022]
Abstract
AIM Histamine receptor H3 (HRH3) has substantial neuropharmacological potential. Currently, knockout models of this receptor have been investigated only in mice. We characterized the expression of this receptor in the zebrafish and generated a zebrafish HRH3 knockout line. Using this model, we studied the role of HRH3 in important behaviours. We also analysed the effect of HRH3 knockout on monoaminergic systems, which has not been thoroughly studied in any animal model. METHODS Generation of a mutant zebrafish line using the CRISPR-Cas9 system. Analysis of locomotor and social behaviour. Expression of HRH3 was characterized using in situ hybridization. Analysis of monoamine networks using HPLC, immunohistochemistry and quantitative PCR. RESULTS We found that HRH3 knockout zebrafish larvae showed a shorter period of increased locomotion after a sudden onset of darkness, while the knockout larvae had a wild-type-like acute response to sudden darkness. Adult knockout fish showed decreased swimming velocity, although locomotor activity of knockout larvae was unaltered. Additionally, levels of dopamine and serotonin were significantly decreased in the knockout fish, while monoamine-related gene expression and immunohistochemistry patterns were unchanged. CONCLUSIONS Our results show that HRH3 knockout larvae adapt faster to sudden darkness, suggesting a role for this receptor in regulating responses to changes in the environment. The decreased levels of dopamine and serotonin provide the first direct evidence that knockout of HRH3 alters these systems.
Collapse
Affiliation(s)
- H. A. J. Puttonen
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| | - M. Sundvik
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| | - S. Semenova
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| | - Y. Shirai
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| | - Y-C. Chen
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| | - P. Panula
- Neuroscience Center and Department of Anatomy; University of Helsinki; Helsinki Finland
| |
Collapse
|
34
|
Vitrac C, Benoit-Marand M. Monoaminergic Modulation of Motor Cortex Function. Front Neural Circuits 2017; 11:72. [PMID: 29062274 PMCID: PMC5640772 DOI: 10.3389/fncir.2017.00072] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/19/2017] [Indexed: 01/09/2023] Open
Abstract
Elaboration of appropriate responses to behavioral situations rests on the ability of selecting appropriate motor outcomes in accordance to specific environmental inputs. To this end, the primary motor cortex (M1) is a key structure for the control of voluntary movements and motor skills learning. Subcortical loops regulate the activity of the motor cortex and thus contribute to the selection of appropriate motor plans. Monoamines are key mediators of arousal, attention and motivation. Their firing pattern enables a direct encoding of different states thus promoting or repressing the selection of actions adapted to the behavioral context. Monoaminergic modulation of motor systems has been extensively studied in subcortical circuits. Despite evidence of converging projections of multiple neurotransmitters systems in the motor cortex pointing to a direct modulation of local circuits, their contribution to the execution and learning of motor skills is still poorly understood. Monoaminergic dysregulation leads to impaired plasticity and motor function in several neurological and psychiatric conditions, thus it is critical to better understand how monoamines modulate neural activity in the motor cortex. This review aims to provide an update of our current understanding on the monoaminergic modulation of the motor cortex with an emphasis on motor skill learning and execution under physiological conditions.
Collapse
Affiliation(s)
- Clément Vitrac
- Laboratoire de Neurosciences Expérimentales et Cliniques, INSERM U1084, Poitiers, France.,Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France
| | - Marianne Benoit-Marand
- Laboratoire de Neurosciences Expérimentales et Cliniques, INSERM U1084, Poitiers, France.,Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, Poitiers, France
| |
Collapse
|
35
|
Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik CM, Hof PR. Monoaminergic neuropathology in Alzheimer's disease. Prog Neurobiol 2017; 151:101-138. [PMID: 27084356 PMCID: PMC5061605 DOI: 10.1016/j.pneurobio.2016.04.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 01/02/2023]
Abstract
None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid β as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, Lille, France
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Claude M Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
36
|
Thr105Ile (rs11558538) polymorphism in the histamine-1-methyl-transferase (HNMT) gene and risk for restless legs syndrome. J Neural Transm (Vienna) 2016; 124:285-291. [DOI: 10.1007/s00702-016-1645-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
|
37
|
Millan MJ, Rivet JM, Gobert A. The frontal cortex as a network hub controlling mood and cognition: Probing its neurochemical substrates for improved therapy of psychiatric and neurological disorders. J Psychopharmacol 2016; 30:1099-1128. [PMID: 27756833 DOI: 10.1177/0269881116672342] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The highly-interconnected and neurochemically-rich frontal cortex plays a crucial role in the regulation of mood and cognition, domains disrupted in depression and other central nervous system disorders, and it is an important site of action for their therapeutic control. For improving our understanding of the function and dysfunction of the frontal cortex, and for identifying improved treatments, quantification of extracellular pools of neuromodulators by microdialysis in freely-moving rodents has proven indispensable. This approach has revealed a complex mesh of autoreceptor and heteroceptor interactions amongst monoaminergic pathways, and led from selective 5-HT reuptake inhibitors to novel classes of multi-target drugs for treating depression like the mixed α2-adrenoceptor/5-HT reuptake inhibitor, S35966, and the clinically-launched vortioxetine and vilazodone. Moreover, integration of non-monoaminergic actions resulted in the discovery and development of the innovative melatonin receptor agonist/5-HT2C receptor antagonist, Agomelatine. Melatonin levels, like those of corticosterone and the "social hormone", oxytocin, can now be quantified by microdialysis over the full 24 h daily cycle. Further, the introduction of procedures for measuring extracellular histamine and acetylcholine has provided insights into strategies for improving cognition by, for example, blockade of 5-HT6 and/or dopamine D3 receptors. The challenge of concurrently determining extracellular levels of GABA, glutamate, d-serine, glycine, kynurenate and other amino acids, and of clarifying their interactions with monoamines, has also been resolved. This has proven important for characterizing the actions of glycine reuptake inhibitors that indirectly augment transmission at N-methyl-d-aspartate receptors, and of "glutamatergic antidepressants" like ketamine, mGluR5 antagonists and positive modulators of AMPA receptors (including S47445). Most recently, quantification of the neurotoxic proteins Aβ42 and Tau has extended microdialysis studies to the pathogenesis of neurodegenerative disorders, and another frontier currently being broached is microRNAs. The present article discusses the above themes, focusses on recent advances, highlights opportunities for clinical "translation", and suggests avenues for further progress.
Collapse
Affiliation(s)
- Mark J Millan
- Pole for Therapeutic Innovation in CNS disorders, IDR Servier, Croissy-sur-Seine, France
| | - Jean-Michel Rivet
- Pole for Therapeutic Innovation in CNS disorders, IDR Servier, Croissy-sur-Seine, France
| | - Alain Gobert
- Pole for Therapeutic Innovation in CNS disorders, IDR Servier, Croissy-sur-Seine, France
| |
Collapse
|
38
|
Maillet A, Krack P, Lhommée E, Météreau E, Klinger H, Favre E, Le Bars D, Schmitt E, Bichon A, Pelissier P, Fraix V, Castrioto A, Sgambato-Faure V, Broussolle E, Tremblay L, Thobois S. The prominent role of serotonergic degeneration in apathy, anxiety and depression inde novoParkinson’s disease. Brain 2016; 139:2486-502. [DOI: 10.1093/brain/aww162] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 05/27/2016] [Indexed: 01/09/2023] Open
|
39
|
Ramsay RR. Molecular aspects of monoamine oxidase B. Prog Neuropsychopharmacol Biol Psychiatry 2016; 69:81-9. [PMID: 26891670 DOI: 10.1016/j.pnpbp.2016.02.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/06/2016] [Accepted: 02/11/2016] [Indexed: 02/07/2023]
Abstract
Monoamine oxidases (MAO) influence the monoamine levels in brain by virtue of their role in neurotransmitter breakdown. MAO B is the predominant form in glial cells and in platelets. MAO B structure, function and kinetics are described as a background for the effect of alterations in its activity on behavior. The need to inhibit MAO B to combat decreased brain amines continues to drive the search for new drugs. Reversible and irreversible inhibitors are now designed using data-mining, computational screening, docking and molecular dynamics. Multi-target ligands designed to combat the elevated activity of MAO B in Alzheimer's and Parkinson's Diseases incorporate MAO inhibition (usually irreversible) as well as iron chelation, antioxidant or neuroprotective properties. The main focus of drug design is the catalytic activity of MAO, but the imidazoline I2 site in the entrance cavity of MAO B is also a pharmacological target. Endogenous regulation of MAO B expression is discussed briefly in light of new studies measuring mRNA, protein, or activity in healthy and degenerative samples, including the effect of DNA methylation on the expression. Overall, this review focuses on examples of recent research on the molecular aspects of the expression, activity, and inhibition of MAO B.
Collapse
Affiliation(s)
- Rona R Ramsay
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews KY16 9ST, United Kingdom.
| |
Collapse
|
40
|
Amini-Khoei H, Rahimi-Balaei M, Amiri S, Haj-Mirzaian A, Hassanipour M, Shirzadian A, Gooshe M, Alijanpour S, Mehr SE, Dehpour AR. Morphine modulates the effects of histamine H1 and H3 receptors on seizure susceptibility in pentylenetetrazole-induced seizure model of mice. Eur J Pharmacol 2015; 769:43-7. [DOI: 10.1016/j.ejphar.2015.10.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 10/07/2015] [Accepted: 10/20/2015] [Indexed: 01/17/2023]
|