1
|
Ren L, Fan Y, Wu W, Qian Y, He M, Li X, Wang Y, Yang Y, Wen X, Zhang R, Li C, Chen X, Hu J. Anxiety disorders: Treatments, models, and circuitry mechanisms. Eur J Pharmacol 2024; 983:176994. [PMID: 39271040 DOI: 10.1016/j.ejphar.2024.176994] [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: 06/09/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
Anxiety disorders are one of the most prevalent mental health conditions worldwide, imposing a significant burden on individuals affected by them and society in general. Current research endeavors aim to enhance the effectiveness of existing anxiolytic drugs and reduce their side effects through optimization or the development of new treatments. Several anxiolytic novel drugs have been produced as a result of discovery-focused research. However, many drug candidates that show promise in preclinical rodent model studies fail to offer any substantive clinical benefits to patients. This review provides an overview of the diagnosis and classification of anxiety disorders together with a systematic review of anxiolytic drugs with a focus on their targets, therapeutic applications, and side effects. It also provides a concise overview of the constraints and disadvantages associated with frequently administered anxiolytic drugs. Additionally, the study comprehensively reviews animal models used in anxiety studies and their associated molecular mechanisms, while also summarizing the brain circuitry related to anxiety. In conclusion, this article provides a valuable foundation for future anxiolytic drug discovery efforts.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China.
| | - Yue Fan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Wenjian Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yuanxin Qian
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Miao He
- College of Life Sciences and Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xinlong Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yizhu Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yu Yang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xuetong Wen
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Ruijia Zhang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Chenhang Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xin Chen
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Jingqing Hu
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Jin F, Lin YY, Wang RC, Xie TX, Zhao Y, Shen C, Sheng D, Ichikawa M, Yu Y, Wang J, Hattori M. Cryo-EM structure of the zinc-activated channel (ZAC) in the Cys-loop receptor superfamily. Proc Natl Acad Sci U S A 2024; 121:e2405659121. [PMID: 39441630 PMCID: PMC11536092 DOI: 10.1073/pnas.2405659121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Cys-loop receptors are a large superfamily of pentameric ligand-gated ion channels with various physiological roles, especially in neurotransmission in the central nervous system. Among them, zinc-activated channel (ZAC) is a Zn2+-activated ion channel that is widely expressed in the human body and is conserved among eukaryotes. Due to its gating by extracellular Zn2+, ZAC has been considered a Zn2+ sensor, but it has undergone minimal structural and functional characterization since its molecular cloning. Among the families in the Cys-loop receptor superfamily, only the structure of ZAC has yet to be determined. Here, we determined the cryo-EM structure of ZAC in the apo state and performed structure-based mutation analyses. We identified a few residues in the extracellular domain whose mutations had a mild impact on Zn2+ sensitivity. The constriction site in the ion-conducting pore differs from the one in other Cys-loop receptor structures, and further mutational analysis identified a key residue that is important for ion selectivity. In summary, our work provides a structural framework for understanding the ion-conducting mechanism of ZAC.
Collapse
Affiliation(s)
- Fei Jin
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Yi-Yu Lin
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Ru-Chun Wang
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Tang-Xuan Xie
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
- Human Phenome Institute, Fudan University, Shanghai201203, China
| | - Cheng Shen
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Danqi Sheng
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Muneyoshi Ichikawa
- State Key Laboratory of Genetic Engineering, Department of Biochemistry and Biophysics, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Ye Yu
- Department of Basic Medicine, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing200098, China
| | - Jin Wang
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing200098, China
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai200438, China
| |
Collapse
|
3
|
Zheng X, Liu G, Ozoe Y, Ju XL. Mechanistic insights into the selectivity of bicyclophosphorothionate antagonists for housefly versus rat GABA receptors. PEST MANAGEMENT SCIENCE 2024; 80:1382-1399. [PMID: 37926485 DOI: 10.1002/ps.7869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/18/2023] [Accepted: 11/06/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND γ-Aminobutyric acid (GABA) receptors (GABARs) are validated targets of insecticides. Bicyclophosphorothionates are a group of insecticidal compounds that act as noncompetitive antagonists of GABARs. We previously reported that the analogs exhibit various degrees of selectivity for housefly versus rat GABARs, depending on substitutions at the 3- and 4-positions. We here sought to elucidate the unsolved mechanisms of the receptor selectivity using quantitative structure-activity relationship (QSAR), molecular docking, and molecular dynamics approaches. RESULTS Three-dimensional (3D)-QSAR studies using Topomer comparative molecular field analysis quantitatively demonstrated how the introduction of a small alkyl group at the 3-position of bicyclophosphorothionates contributes to the housefly versus rat GABAR selectivity. To investigate the molecular mechanisms of the selective action, bicyclophosphorothionates were docked into housefly Resistance to dieldrin (RDL) GABAR and rat α1β2γ2 GABAR homology models built using the published 3D-structures of human GABARs as templates. The results of molecular docking and molecular dynamics simulations revealed that the 2'Ala and 6'Thr residues of the RDL subunit within the channel are the key amino acids for binding to the housefly GABARs, whereas the 2'Ser residue of γ2 subunit plays a crucial role in binding to rat GABARs. CONCLUSION We revealed the molecular mechanisms underlying the selective antagonistic action of bicyclophosphorothionates on housefly versus rat GABARs. The information presented should help design and develop novel, safe GABAR-targeting insecticides. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaojiao Zheng
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Genyan Liu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| | - Yoshihisa Ozoe
- Department of Life Science and Biotechnology, Faculty of Life and Environmental Sciences, Shimane University, Matsue, Japan
| | - Xiu-Lian Ju
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, P. R. China
| |
Collapse
|
4
|
Tipton AE, Russek SJ. Regulation of Inhibitory Signaling at the Receptor and Cellular Level; Advances in Our Understanding of GABAergic Neurotransmission and the Mechanisms by Which It Is Disrupted in Epilepsy. Front Synaptic Neurosci 2022; 14:914374. [PMID: 35874848 PMCID: PMC9302637 DOI: 10.3389/fnsyn.2022.914374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Inhibitory signaling in the brain organizes the neural circuits that orchestrate how living creatures interact with the world around them and how they build representations of objects and ideas. Without tight control at multiple points of cellular engagement, the brain’s inhibitory systems would run down and the ability to extract meaningful information from excitatory events would be lost leaving behind a system vulnerable to seizures and to cognitive decline. In this review, we will cover many of the salient features that have emerged regarding the dynamic regulation of inhibitory signaling seen through the lens of cell biology with an emphasis on the major building blocks, the ligand-gated ion channel receptors that are the first transduction point when the neurotransmitter GABA is released into the synapse. Epilepsy association will be used to indicate importance of key proteins and their pathways to brain function and to introduce novel areas for therapeutic intervention.
Collapse
Affiliation(s)
- Allison E. Tipton
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
| | - Shelley J. Russek
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
- *Correspondence: Shelley J. Russek,
| |
Collapse
|
5
|
Niu L, Hou Y, Jiang M, Bai G. The rich pharmacological activities of Magnolia officinalis and secondary effects based on significant intestinal contributions. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114524. [PMID: 34400262 DOI: 10.1016/j.jep.2021.114524] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/01/2021] [Accepted: 08/13/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis Cortex (M. officinalis) is a traditional herbal drug widely used in Asian countries. Depending on its multiple biological activities, M. officinalis is used to regulate gastrointestinal (GI) motility, relieve cough and asthma, prevent cardiovascular and cerebrovascular diseases, and treat depression and anxiety. AIM OF THE REVIEW We aimed to review the abundant form of pharmacodynamics activity and potential mechanisms of action of M. officinalis and the characteristics of the internal processes of the main components. The potential mechanisms of local and distance actions of M. officinalis based on GI tract was provided, and it was used to reveal the interconnections between traditional use, phytochemistry, and pharmacology. MATERIALS AND METHODS Published literatures about M. officinalis and its main components were collected from several scientific databases, including PubMed, Elsevier, ScienceDirect, Google Scholar and Web of Science etc. RESULTS: M. officinalis was shown multiple effects including effects on digestive system, respiratory system, central system, which is consistent with traditional applications, as well as some other activities such as cardiovascular system, anticancer, anti-inflammatory and antioxidant effects and so on. The mechanisms of these activities are abundant. Its chief ingredients such as magnolol and honokiol can be metabolized into active metabolites in vivo, which can increase water solubility and bioavailability and exert pharmacological activity in the whole body. In the GI tract, M. officinalis and its main ingredient can regulate GI hormones and substance metabolism, protect the intestinal barrier and affect the gut microbiota (GM). These actions are effective to improve local discomfort and some distal symptoms such as depression, asthma, or metabolic disorders. CONCLUSIONS Although M. officinalis has rich pharmacological effects, the GI tract makes great contributions to it. The GI tract is not only an important place for absorption and metabolism but also a key site to help M. officinalis exert local and distal efficacy. Pharmacodynamical studies on the efficacies of distal tissues based on the contributions of the GI tract hold great potential for understanding the benefits of M. officinalis and providing new ideas for the treatment of important diseases.
Collapse
Affiliation(s)
- Lin Niu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, People's Republic of China.
| |
Collapse
|
6
|
Effect of Garlic ( Allium sativum) Supplementation on Premenstrual Disorders: A Randomized, Double-Blind, Placebo-Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9965064. [PMID: 34760020 PMCID: PMC8575623 DOI: 10.1155/2021/9965064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 10/07/2021] [Indexed: 11/22/2022]
Abstract
Background Premenstrual disorders involve physical, behavioral, and mood variations that affect women of childbearing age and interfere with family relationships, household responsibilities, professional duties, and social activities. Objectives Considering the side effects of conventional medications, their use is not recommended except in severe cases of premenstrual disorders. Nowadays, there is a tendency to use traditional and complementary medicine that offers various treatments. The purpose of the current study was to investigate the impacts of garlic as a herbal medicine on the severity of premenstrual symptoms. Methods This study was a double-blind, randomized, controlled trial. After identification of participants with moderate-to-severe PMS through the premenstrual symptoms screening tools questionnaire (PSST), they were randomly assigned to placebo (n = 64) or garlic (n = 65) groups. Each participant received one tablet daily for three consecutive cycles and logged the severity of their symptoms in the PSST questionnaire during the intervention period. Results There was no significant difference between the two groups in the baseline level of premenstrual symptoms before the intervention. After treatment with garlic for three consecutive cycles, the total score of the severity of premenstrual symptoms significantly (P < 0.001) reduced from 34.09 ± 7.31 to 11.21 ± 7.17. In the placebo group, this score changed from 33.35 ± 7.96 to 24.28 ± 7.22. The difference between mean changes in the two groups was 13.78, with a 95% Confidence Interval (CI) of 11.23–16.33. No serious side effects were observed in either group. Conclusion Our findings highlight the potential effect of garlic in reducing the severity of premenstrual symptoms; therefore, the use of garlic can be considered as an alternative therapy in the prevention and treatment of premenstrual disorders.
Collapse
|
7
|
Electrophysiology of ionotropic GABA receptors. Cell Mol Life Sci 2021; 78:5341-5370. [PMID: 34061215 PMCID: PMC8257536 DOI: 10.1007/s00018-021-03846-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/02/2021] [Accepted: 04/23/2021] [Indexed: 10/30/2022]
Abstract
GABAA receptors are ligand-gated chloride channels and ionotropic receptors of GABA, the main inhibitory neurotransmitter in vertebrates. In this review, we discuss the major and diverse roles GABAA receptors play in the regulation of neuronal communication and the functioning of the brain. GABAA receptors have complex electrophysiological properties that enable them to mediate different types of currents such as phasic and tonic inhibitory currents. Their activity is finely regulated by membrane voltage, phosphorylation and several ions. GABAA receptors are pentameric and are assembled from a diverse set of subunits. They are subdivided into numerous subtypes, which differ widely in expression patterns, distribution and electrical activity. Substantial variations in macroscopic neural behavior can emerge from minor differences in structure and molecular activity between subtypes. Therefore, the diversity of GABAA receptors widens the neuronal repertoire of responses to external signals and contributes to shaping the electrical activity of neurons and other cell types.
Collapse
|
8
|
Molecular Targets of Cannabidiol in Experimental Models of Neurological Disease. Molecules 2020; 25:molecules25215186. [PMID: 33171772 PMCID: PMC7664437 DOI: 10.3390/molecules25215186] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Cannabidiol (CBD) is a non-psychoactive phytocannabinoid known for its beneficial effects including antioxidant and anti-inflammatory properties. Moreover, CBD is a compound with antidepressant, anxiolytic, anticonvulsant and antipsychotic effects. Thanks to all these properties, the interest of the scientific community for it has grown. Indeed, CBD is a great candidate for the management of neurological diseases. The purpose of our review is to summarize the in vitro and in vivo studies published in the last 15 years that describe the biochemical and molecular mechanisms underlying the effects of CBD and its therapeutic application in neurological diseases. CBD exerts its neuroprotective effects through three G protein coupled-receptors (adenosine receptor subtype 2A, serotonin receptor subtype 1A and G protein-coupled receptor 55), one ligand-gated ion channel (transient receptor potential vanilloid channel-1) and one nuclear factor (peroxisome proliferator-activated receptor γ). Moreover, the therapeutical properties of CBD are also due to GABAergic modulation. In conclusion, CBD, through multi-target mechanisms, represents a valid therapeutic tool for the management of epilepsy, Alzheimer’s disease, multiple sclerosis and Parkinson’s disease.
Collapse
|
9
|
Kim DH, Kwon H, Choi JW, Shin CY, Cheong JH, Park SJ, Ryu JH. Roles of GABA A receptor α5 subunit on locomotion and working memory in transient forebrain ischemia in mice. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109962. [PMID: 32428535 DOI: 10.1016/j.pnpbp.2020.109962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
The γ-aminobutyric acid A (GABAA) receptor, which contains a chloride channel, is a typical inhibitory neurotransmitter receptor in the central nervous system. Although the GABAergic neurotransmitter system has been discovered to be involved in various psychological behaviors, such as anxiety, convulsions, and cognitive function, its functional changes under conditions of ischemic pathological situation are still uncovered. In the present study, we attempted to elucidate the functional changes in the GABAergic system after transient forebrain ischemia in mice. A bilateral common carotid artery occlusion for 20 min was used to establish a model of transient forebrain ischemia/reperfusion (tI/R). Delayed treatment with diazepam, a positive allosteric modulator of the GABAA receptor, increased locomotor activity in the open field test and spontaneous alternations in the Y-maze test in tI/R mice, but not in shams. Delayed treatment with diazepam did not alter neuronal death or the number of GABAergic neurons in tI/R mice. However, tI/R induced changes in the protein levels of GABAA receptor subunits in the hippocampus. In particular, the most marked increase in the tI/R group was found in the level of α5 subunit of the GABAA receptor. Similar to delayed treatment with diazepam, delayed treatment with imidazenil, an α5-sensitive benzodiazepine, increased spontaneous alternations in the Y-maze in tI/R mice, whereas zolpidem, an α5-insensitive benzodiazepine, failed to show such effects. These results suggest that tI/R-induced changes in the level of the α5 subunit of the GABAA receptor can alter the function of GABAergic drugs in a mouse model of forebrain ischemia.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea; Institute of Convergence Bio-Health, Dong-A University, Busan 49315, Republic of Korea.
| | - Huiyoung Kwon
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan 49315, Republic of Korea.
| | - Ji Woong Choi
- Laboratory of Neuropharmacology, College of Pharmacy, Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, Republic of Korea.
| | - Chan Young Shin
- Departments of Pharmacology and Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea.
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea.
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Jong Hoon Ryu
- Department of Life and Nanopharmaceutical Sciences, Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
10
|
Li C, Lei Y, Tian Y, Xu S, Shen X, Wu H, Bao S, Wang F. The etiological contribution of GABAergic plasticity to the pathogenesis of neuropathic pain. Mol Pain 2020; 15:1744806919847366. [PMID: 30977423 PMCID: PMC6509976 DOI: 10.1177/1744806919847366] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuropathic pain developing after peripheral or central nerve injury is the result of pathological changes generated through complex mechanisms. Disruption in the homeostasis of excitatory and inhibitory neurons within the central nervous system is a crucial factor in the formation of hyperalgesia or allodynia occurring with neuropathic pain. The central GABAergic pathway has received attention for its extensive distribution and function in neural circuits, including the generation and development of neuropathic pain. GABAergic inhibitory changes that occur in the interneurons along descending modulatory and nociceptive pathways in the central nervous system are believed to generate neuronal plasticity, such as synaptic plasticity or functional plasticity of the related genes or proteins, that is the foundation of persistent neuropathic pain. The primary GABAergic plasticity observed in neuropathic pain includes GABAergic synapse homo- and heterosynaptic plasticity, decreased synthesis of GABA, down-expression of glutamic acid decarboxylase and GABA transporter, abnormal expression of NKCC1 or KCC2, and disturbed function of GABA receptors. In this review, we describe possible mechanisms associated with GABAergic plasticity, such as central sensitization and GABAergic interneuron apoptosis, and the epigenetic etiologies of GABAergic plasticity in neuropathic pain. Moreover, we summarize potential therapeutic targets of GABAergic plasticity that may allow for successful relief of hyperalgesia from nerve injury. Finally, we compare the effects of the GABAergic system in neuropathic pain to other types of chronic pain to understand the contribution of GABAergic plasticity to neuropathic pain.
Collapse
Affiliation(s)
- Caijuan Li
- 1 Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yanying Lei
- 2 Department of Stomatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yi Tian
- 3 Department of Anesthesiology, Haikou Affiliated Hospital of Xiangya Medical School, Central South University, Haikou People's Hospital, Haikou, China
| | - Shiqin Xu
- 1 Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiaofeng Shen
- 1 Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Haibo Wu
- 1 Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Senzhu Bao
- 2 Department of Stomatology, Affiliated Hospital of Qinghai University, Xining, China
| | - Fuzhou Wang
- 1 Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.,4 Group of Neuropharmacology and Neurophysiology, Division of Neuroscience, The Bonoi Academy of Science and Education, Chapel Hill, NC, USA
| |
Collapse
|
11
|
Hasselmo ME, Alexander AS, Hoyland A, Robinson JC, Bezaire MJ, Chapman GW, Saudargiene A, Carstensen LC, Dannenberg H. The Unexplored Territory of Neural Models: Potential Guides for Exploring the Function of Metabotropic Neuromodulation. Neuroscience 2020; 456:143-158. [PMID: 32278058 DOI: 10.1016/j.neuroscience.2020.03.048] [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: 12/10/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022]
Abstract
The space of possible neural models is enormous and under-explored. Single cell computational neuroscience models account for a range of dynamical properties of membrane potential, but typically do not address network function. In contrast, most models focused on network function address the dimensions of excitatory weight matrices and firing thresholds without addressing the complexities of metabotropic receptor effects on intrinsic properties. There are many under-explored dimensions of neural parameter space, and the field needs a framework for representing what has been explored and what has not. Possible frameworks include maps of parameter spaces, or efforts to categorize the fundamental elements and molecules of neural circuit function. Here we review dimensions that are under-explored in network models that include the metabotropic modulation of synaptic plasticity and presynaptic inhibition, spike frequency adaptation due to calcium-dependent potassium currents, and afterdepolarization due to calcium-sensitive non-specific cation currents and hyperpolarization activated cation currents. Neuroscience research should more effectively explore possible functional models incorporating under-explored dimensions of neural function.
Collapse
Affiliation(s)
- Michael E Hasselmo
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States.
| | - Andrew S Alexander
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Alec Hoyland
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Jennifer C Robinson
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Marianne J Bezaire
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - G William Chapman
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Ausra Saudargiene
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Lucas C Carstensen
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| | - Holger Dannenberg
- Center for Systems Neuroscience, Department of Psychological and Brain Sciences, Boston University, 610 Commonwealth Ave., Boston, MA 02215, United States
| |
Collapse
|
12
|
Yim PD, Gallos G, Lee-Kong SA, Dan W, Wu AD, Xu D, Berkowitz DE, Emala CW. Novel Expression of GABAA Receptors on Resistance Arteries That Modulate Myogenic Tone. J Vasc Res 2020; 57:113-125. [PMID: 32097943 DOI: 10.1159/000505456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/16/2019] [Indexed: 01/17/2023] Open
Abstract
The clinical administration of GABAergic medications leads to hypotension which has classically been attributed to the modulation of neuronal activity in the central and peripheral nervous systems. However, certain types of peripheral smooth muscle cells have been shown to express GABAA receptors, which modulate smooth muscle tone, by the activation of these chloride channels on smooth muscle cell plasma membranes. Limited prior studies demonstrate that non-human large-caliber capacitance blood vessels mounted on a wire myograph are responsive to GABAA ligands. We questioned whether GABAA receptors are expressed in human resistance arteries and whether they modulate myogenic tone. We demonstrate the novel expression of GABAA subunits on vascular smooth muscle from small-caliber human omental and mouse tail resistance arteries. We show that GABAA receptors modulate both plasma membrane potential and calcium responses in primary cultured cells from human resistance arteries. Lastly, we demonstrate functional physiologic modulation of myogenic tone via GABAA receptor activation in human and mouse arteries. Together, these studies demonstrate a previously unrecognized role for GABAA receptors in the modulation of myogenic tone in mouse and human resistance arteries.
Collapse
Affiliation(s)
- Peter D Yim
- Department of Anesthesiology, Columbia University, New York, New York, USA,
| | - George Gallos
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | | | - William Dan
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | - Amy D Wu
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | - Dingbang Xu
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | - Dan E Berkowitz
- Department of Anesthesiology and Perioperative Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Charles W Emala
- Department of Anesthesiology, Columbia University, New York, New York, USA
| |
Collapse
|
13
|
Cifelli P, Ruffolo G, De Felice E, Alfano V, van Vliet EA, Aronica E, Palma E. Phytocannabinoids in Neurological Diseases: Could They Restore a Physiological GABAergic Transmission? Int J Mol Sci 2020; 21:E723. [PMID: 31979108 PMCID: PMC7038116 DOI: 10.3390/ijms21030723] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 01/19/2023] Open
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are the main inhibitory mediators in the central nervous system (CNS). GABAARs are pentameric ligand gated ion channels, and the main subunit composition is usually 2α2βγ, with various isotypes assembled within a set of 19 different subunits. The inhibitory function is mediated by chloride ion movement across the GABAARs, activated by synaptic GABA release, reducing neuronal excitability in the adult CNS. Several studies highlighted the importance of GABA-mediated transmission during neuro-development, and its involvement in different neurological and neurodevelopmental diseases, from anxiety to epilepsy. However, while it is well known how different classes of drugs are able to modulate the GABAARs function (benzodiazepines, barbiturates, neurosteroids, alcohol), up to now little is known about GABAARs and cannabinoids interaction in the CNS. Endocannabinoids and phytocannabinoids are lately emerging as a new class of promising drugs for a wide range of neurological conditions, but their safety as medication, and their mechanisms of action are still to be fully elucidated. In this review, we will focus our attention on two of the most promising molecules (Δ9-tetrahydrocannabinol; Δ9-THC and cannabidiol; CBD) of this new class of drugs and their possible mechanism of action on GABAARs.
Collapse
Affiliation(s)
| | - Gabriele Ruffolo
- IRCCS San Raffaele Pisana, 00163 Rome, Italy; (G.R.); (E.D.F.)
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, 00185 Rome, Italy;
| | | | - Veronica Alfano
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, 00185 Rome, Italy;
| | - Erwin Alexander van Vliet
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, 1105 Amsterdam, The Netherlands; (E.A.v.V.); (E.A.)
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1090 Amsterdam, The Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, 1105 Amsterdam, The Netherlands; (E.A.v.V.); (E.A.)
- Stichting Epilepsie Instellingen Nederland (SEIN), 0397 Heemstede, The Netherlands
| | - Eleonora Palma
- Department of Physiology and Pharmacology, laboratory affiliated to Istituto Pasteur Italia, University of Rome Sapienza, 00185 Rome, Italy;
| |
Collapse
|
14
|
Li C, Li Y, Zhao Z, Lv Y, Gu B, Zhao L. Aerobic exercise regulates synaptic transmission and reactive oxygen species production in the paraventricular nucleus of spontaneously hypertensive rats. Brain Res 2019; 1712:82-92. [PMID: 30735639 DOI: 10.1016/j.brainres.2019.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023]
Abstract
Aerobic exercise lowers blood pressure in patients with hypertension, but the underlying mechanisms remain incompletely understood. The hypothalamic paraventricular nucleus (PVN) plays a key role in the control of sympathetic outflow and cardiovascular tone. We examined whether chronic aerobic exercise altered synaptic transmission and reactive oxygen species (ROS) production in the PVN. In the present study, spontaneously hypertensive rats (SHRs) were subjected to exercise training for 8 weeks, five times per week, with Wistar Kyoto (WKY) rats as the cohort control. Miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs) were recorded from the PVN in ex vivo hypothalamic slice preparations obtained after the last training, and biomarkers of oxidative stress and physical indexes were observed. The mean frequency and amplitude, as well as the rise time and the decay time constant of mIPSCs, significantly decreased in 20-wk-old SHRs compared to WKY 20-wk-old controls. In contrast to mIPSCs, only the mean mEPSC frequency was higher, and there were no other changes in mEPSCs in comparison to the control group. SHRs exhibited higher ROS, 8-OHdG, and MDA; and lower SOD1, SOD2, CAT, Ogg1, and SOD and CAT activity in the PVN. These SHRs also had a significant increase in heart rate, blood pressure and sympathetic nerve activity, and higher levels of norepinephrine (NE). Exercise training ameliorated all these abnormalities, resulting in an increase in the mean frequency, amplitude and kinetics of mIPSCs, accompanied by a decrease in the mean frequency of mEPSCs in the PVN. This study demonstrates that moderate intensity, high frequency exercise training induces a selective enhancement of inhibitory synaptic transmission in the PVN, which may dampen sympathetic activity and reduce blood pressure in hypertension. These changes may be due to antioxidant-related adaptations in the PVNs of SHRs.
Collapse
Affiliation(s)
- Cui Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Ziqi Zhao
- College of Life Science, University of Chinese Academy of Science, Beijing, China
| | - Yuanyuan Lv
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| | - Li Zhao
- Department of Exercise Physiology, Beijing Sport University, Beijing, China; Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China.
| |
Collapse
|
15
|
Li Z, Cogswell M, Hixson K, Brooks-Kayal AR, Russek SJ. Nuclear Respiratory Factor 1 (NRF-1) Controls the Activity Dependent Transcription of the GABA-A Receptor Beta 1 Subunit Gene in Neurons. Front Mol Neurosci 2018; 11:285. [PMID: 30186109 PMCID: PMC6113564 DOI: 10.3389/fnmol.2018.00285] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/27/2018] [Indexed: 11/13/2022] Open
Abstract
While the exact role of β1 subunit-containing GABA-A receptors (GABARs) in brain function is not well understood, altered expression of the β1 subunit gene (GABRB1) is associated with neurological and neuropsychiatric disorders. In particular, down-regulation of β1 subunit levels is observed in brains of patients with epilepsy, autism, bipolar disorder and schizophrenia. A pathophysiological feature of these disease states is imbalance in energy metabolism and mitochondrial dysfunction. The transcription factor, nuclear respiratory factor 1 (NRF-1), has been shown to be a key mediator of genes involved in oxidative phosphorylation and mitochondrial biogenesis. Using a variety of molecular approaches (including mobility shift, promoter/reporter assays, and overexpression of dominant negative NRF-1), we now report that NRF-1 regulates transcription of GABRB1 and that its core promoter contains a conserved canonical NRF-1 element responsible for sequence specific binding and transcriptional activation. Our identification of GABRB1 as a new target for NRF-1 in neurons suggests that genes coding for inhibitory neurotransmission may be coupled to cellular metabolism. This is especially meaningful as binding of NRF-1 to its element is sensitive to the kind of epigenetic changes that occur in multiple disorders associated with altered brain inhibition.
Collapse
Affiliation(s)
- Zhuting Li
- Laboratory of Translational Epilepsy, Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States.,Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Meaghan Cogswell
- Laboratory of Translational Epilepsy, Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Kathryn Hixson
- Laboratory of Translational Epilepsy, Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States
| | - Amy R Brooks-Kayal
- Department of Pediatrics, Division of Neurology, School of Medicine, University of Colorado, Aurora, CO, United States.,Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Shelley J Russek
- Laboratory of Translational Epilepsy, Department of Pharmacology and Experimental Therapeutics, School of Medicine, Boston University, Boston, MA, United States.,Department of Biology, Boston University, Boston, MA, United States
| |
Collapse
|
16
|
Cicvaric A, Bulat T, Bormann D, Yang J, Auer B, Milenkovic I, Cabatic M, Milicevic R, Monje FJ. Sustained consumption of cocoa-based dark chocolate enhances seizure-like events in the mouse hippocampus. Food Funct 2018; 9:1532-1544. [PMID: 29431797 DOI: 10.1039/c7fo01668a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the consumption of caffeine and cocoa has been associated with a variety of health benefits to humans, some authors have proposed that excessive caffeine intake may increase the frequency of epileptic seizures in humans and reduce the efficiency of antiepileptic drugs. Little is known, however, about the proconvulsant potential of the sustained, excessive intake of cocoa on hippocampal neural circuits. Using the mouse as an experimental model, we examined the effects of the chronic consumption of food enriched in cocoa-based dark chocolate on motor and mood-related behaviours as well as on the excitability properties of hippocampal neurons. Cocoa food enrichment did not affect body weights or mood-related behaviours but rather promoted general locomotion and improved motor coordination. However, ex vivo electrophysiological analysis revealed a significant enhancement in seizure-like population spike bursting at the neurogenic dentate gyrus, which was paralleled by a significant reduction in the levels of GABA-α1 receptors thus suggesting that an excessive dietary intake of cocoa-enriched food might alter some of the synaptic elements involved in epileptogenesis. These data invite further multidisciplinary research aiming to elucidate the potential deleterious effects of chocolate abuse on behaviour and brain hyperexcitability.
Collapse
Affiliation(s)
- Ana Cicvaric
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Tanja Bulat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Daniel Bormann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Jiaye Yang
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Bastian Auer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Ivan Milenkovic
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Maureen Cabatic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Radoslav Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Francisco J Monje
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| |
Collapse
|
17
|
Ding S, Zhuge W, Hu J, Yang J, Wang X, Wen F, Wang C, Zhuge Q. Baicalin reverses the impairment of synaptogenesis induced by dopamine burden via the stimulation of GABA AR-TrkB interaction in minimal hepatic encephalopathy. Psychopharmacology (Berl) 2018; 235:1163-1178. [PMID: 29404643 PMCID: PMC5869945 DOI: 10.1007/s00213-018-4833-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 01/08/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND It has been reported that D1 receptor (D1R) activation reduces GABAA receptor (GABAAR) current, and baicalin (BAI) displays therapeutic efficacy in diseases involving cognitive impairment. METHODS We investigated the mechanisms by which BAI could improve DA-induced minimal hepatic encephalopathy (MHE) using immunoblotting, immunofluorescence, and co-immunoprecipitation. RESULTS BAI did not induce toxicity on the primary cultured neurons. And no obvious toxicity of BAI to the brain was found in rats. DA activated D1R/dopamine and adenosine 3'5'-monophosphate-regulated phospho-protein (DARPP32) to reduce the GABAAR current; BAI treatment did not change the D1R/DARPP32 levels but blocked DA-induced reduction of GABAAR levels in primary cultured neurons. DA decreased the interaction of GABAAR with TrkB and the expression of downstream AKT, which was blocked by BAI treatment. Moreover, BAI reversed the decrease in the expression of GABAAR/TrkB/AKT and prevented the impairment of synaptogenesis and memory deficits in MHE rats. CONCLUSIONS These results suggest that BAI has neuroprotective and synaptoprotective effects on MHE which are not related to upstream D1R/DARPP32 signaling, but to the targeting of downstream GABAAR signaling to TrkB.
Collapse
Affiliation(s)
- Saidan Ding
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Weishan Zhuge
- Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Jiangnan Hu
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107 USA
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Neurosurgery Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Xuebao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Fangfang Wen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Chengde Wang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Neurosurgery Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000 People’s Republic of China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Neurosurgery Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.
| |
Collapse
|
18
|
Ramesh SA, Tyerman SD, Gilliham M, Xu B. γ-Aminobutyric acid (GABA) signalling in plants. Cell Mol Life Sci 2017; 74:1577-1603. [PMID: 27838745 PMCID: PMC11107511 DOI: 10.1007/s00018-016-2415-7] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/06/2016] [Accepted: 11/08/2016] [Indexed: 01/11/2023]
Abstract
The role of γ-aminobutyric acid (GABA) as a signal in animals has been documented for over 60 years. In contrast, evidence that GABA is a signal in plants has only emerged in the last 15 years, and it was not until last year that a mechanism by which this could occur was identified-a plant 'GABA receptor' that inhibits anion passage through the aluminium-activated malate transporter family of proteins (ALMTs). ALMTs are multigenic, expressed in different organs and present on different membranes. We propose GABA regulation of ALMT activity could function as a signal that modulates plant growth, development, and stress response. In this review, we compare and contrast the plant 'GABA receptor' with mammalian GABAA receptors in terms of their molecular identity, predicted topology, mode of action, and signalling roles. We also explore the implications of the discovery that GABA modulates anion flux in plants, its role in signal transduction for the regulation of plant physiology, and predict the possibility that there are other GABA interaction sites in the N termini of ALMT proteins through in silico evolutionary coupling analysis; we also explore the potential interactions between GABA and other signalling molecules.
Collapse
Affiliation(s)
- Sunita A Ramesh
- Plant Transport and Signalling Lab, ARC Centre of Excellence in Plant Energy Biology and School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Glen Osmond, SA, 5064, Australia
| | - Stephen D Tyerman
- Plant Transport and Signalling Lab, ARC Centre of Excellence in Plant Energy Biology and School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Glen Osmond, SA, 5064, Australia
| | - Matthew Gilliham
- Plant Transport and Signalling Lab, ARC Centre of Excellence in Plant Energy Biology and School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Glen Osmond, SA, 5064, Australia
| | - Bo Xu
- Plant Transport and Signalling Lab, ARC Centre of Excellence in Plant Energy Biology and School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Glen Osmond, SA, 5064, Australia.
| |
Collapse
|
19
|
Kim HS, Jin EH, Mo JS, Chae SC. Significant association of the GABRP polymorphisms with ulcerative colitis patients. Genes Genomics 2017. [DOI: 10.1007/s13258-017-0514-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Maldifassi MC, Baur R, Sigel E. Molecular mode of action of CGS 9895 at α1β2γ2GABAAreceptors. J Neurochem 2016; 138:722-30. [DOI: 10.1111/jnc.13711] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/01/2016] [Accepted: 06/04/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Maria C. Maldifassi
- Institute of Biochemistry and Molecular Medicine; University of Bern; Bern Switzerland
| | - Roland Baur
- Institute of Biochemistry and Molecular Medicine; University of Bern; Bern Switzerland
| | - Erwin Sigel
- Institute of Biochemistry and Molecular Medicine; University of Bern; Bern Switzerland
| |
Collapse
|
21
|
Borroni MV, Vallés AS, Barrantes FJ. The lipid habitats of neurotransmitter receptors in brain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2662-2670. [PMID: 27424801 DOI: 10.1016/j.bbamem.2016.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/05/2016] [Accepted: 07/12/2016] [Indexed: 12/17/2022]
Abstract
Neurotransmitter receptors, the macromolecules specialized in decoding the chemical signals encrypted in the chemical signaling mechanism in the nervous system, occur either at the somatic cell surface of chemically excitable cells or at specialized subcellular structures, the synapses. Synapses have lipid compositions distinct from the rest of the cell membrane, suggesting that neurotransmitter receptors and their scaffolding and adaptor protein partners require specific lipid habitats for optimal operation. In this review we discuss some paradigmatic cases of neurotransmitter receptor-lipid interactions, highlighting the chemical nature of the intervening lipid species and providing examples of the receptor mechanisms affected by interaction with lipids. The focus is on the effects of cholesterol, glycerophospholipids and covalent fatty acid acylation on neurotransmitter receptors. We also briefly discuss the role of lipid phase states involving lateral heterogeneities of the host membrane known to modulate membrane transport, protein sorting and signaling. Modulation of neurotransmitter receptors by lipids occurs at multiple levels, affecting a wide span of activities including their trafficking, sorting, stability, residence lifetime at the cell surface, endocytosis, and recycling, among other important functional properties at the synapse.
Collapse
Affiliation(s)
- María Virginia Borroni
- Instituto de Tecnología en Polímeros y Nanotecnología (ITPN) Av. Las Heras 2214 C1127AAQ Buenos Aires Argentina
| | - Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, B8000FWB Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute, UCA-CONICET, Faculty of Medical Sciences, Catholic University of Argentina, Av. Alicia Moreau de Justo 1600, C1107AFF Buenos Aires, Argentina.
| |
Collapse
|
22
|
Maldifassi MC, Baur R, Pierce D, Nourmahnad A, Forman SA, Sigel E. Novel positive allosteric modulators of GABAA receptors with anesthetic activity. Sci Rep 2016; 6:25943. [PMID: 27198062 PMCID: PMC4873749 DOI: 10.1038/srep25943] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 04/25/2016] [Indexed: 01/08/2023] Open
Abstract
GABAA receptors are the main inhibitory neurotransmitter receptors in the brain and are targets for numerous clinically important drugs such as benzodiazepines, anxiolytics and anesthetics. We previously identified novel ligands of the classical benzodiazepine binding pocket in α1β2γ2 GABAA receptors using an experiment-guided virtual screening (EGVS) method. This screen also identified novel ligands for intramembrane low affinity diazepam site(s). In the current study we have further characterized compounds 31 and 132 identified with EGVS as well as 4-O-methylhonokiol. We investigated the site of action of these compounds in α1β2γ2 GABAA receptors expressed in Xenopus laevis oocytes using voltage-clamp electrophysiology combined with a benzodiazepine site antagonist and transmembrane domain mutations. All three compounds act mainly through the two β+/α− subunit transmembrane interfaces of the GABAA receptors. We then used concatenated receptors to dissect the involvement of individual β+/α− interfaces. We further demonstrated that these compounds have anesthetic activity in a small aquatic animal model, Xenopus laevis tadpoles. The newly identified compounds may serve as scaffolds for the development of novel anesthetics.
Collapse
Affiliation(s)
- Maria C Maldifassi
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Roland Baur
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - David Pierce
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, 02114 Massachusetts
| | - Anahita Nourmahnad
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, 02114 Massachusetts
| | - Stuart A Forman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, 02114 Massachusetts
| | - Erwin Sigel
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| |
Collapse
|
23
|
Liao C, Han Q, Ma Y, Su B. Age-related gene expression change of GABAergic system in visual cortex of rhesus macaque. Gene 2016; 590:227-33. [PMID: 27196061 DOI: 10.1016/j.gene.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/03/2016] [Accepted: 05/08/2016] [Indexed: 01/31/2023]
Abstract
Degradation of visual function is a common phenomenon during aging and likely mediated by change in the impaired central visual pathway. Treatment with GABA or its agonist could recover the ability of visual neurons in the primary visual cortex of senescent macaques. However, little is known about how GABAergic system change is related to the aged degradation of visual function in nonhuman primate. With the use of quantitative PCR method, we measured the expression change of 24 GABA related genes in the primary visual cortex (Brodmann's 17) of different age groups. In this study, both of mRNA and protein of glutamic acid decarboxylase (GAD65) were measured by real-time RT-PCR and Western blot, respectively. Results revealed that the level of GAD65 message was not significantly altered, but the proteins were significantly decreased in the aged monkey. As GAD65 plays an important role in GABA synthesis, the down-regulation of GAD65 protein was likely the key factor leading to the observed GABA reduction in the primary visual cortex of the aged macaques. In addition, 7 of 14 GABA receptor genes were up-regulated and one GABA receptor gene was significantly reduced during aging process even after Banjamini correction for multiple comparisons (P<0.05). These results suggested that the dysregulation of GAD65 protein might contribute to some age-related neural visual dysfunctions and most of GABA receptor genes induce a clear indication of compensatory effect for the reduced GABA release in the healthy aged monkey cortex.
Collapse
Affiliation(s)
- Chenghong Liao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, 570228, China; Laboratory of Tropical Veterinary Medicine and Vector Biology, College of Agriculture, Hainan University, Haikou, 570228, China
| | - Qian Han
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, 570228, China; Laboratory of Tropical Veterinary Medicine and Vector Biology, College of Agriculture, Hainan University, Haikou, 570228, China
| | - Yuanye Ma
- Laboratory of the Primate Model for Brain Diseases and Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming 650223, China; State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
24
|
Ruiz-Veguilla M, Martín-Rodríguez JF, Palomar FJ, Porcacchia P, Álvarez de Toledo P, Perona-Garcelán S, Rodríguez-Testal JF, Huertas-Fernández I, Mir P. Trait- and state-dependent cortical inhibitory deficits in bipolar disorder. Bipolar Disord 2016; 18:261-71. [PMID: 27004755 DOI: 10.1111/bdi.12382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/08/2016] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Euthymic patients with bipolar disorder (BD) have deficits in cortical inhibition. However, whether cortical inhibitory deficits are trait- or state-dependent impairments is not yet known and their relationship with psychiatric symptoms is not yet understood. In the present study, we examined trait- and state-dependent cortical inhibitory deficits and evaluated the potential clinical significance of these deficits. METHODS Nineteen patients with bipolar I disorder were evaluated using the paired-pulse transcranial stimulation protocol, which assessed cortical inhibition during an acute manic episode. Cortical inhibition measures were compared with those obtained in 28 demographically matched healthy controls. A follow-up assessment was performed in 15 of these patients three months later, when there was remission from their mood and psychotic symptoms. The association between cortical inhibitory measures and severity of psychiatric symptoms was also studied. RESULTS During mania, patients showed decreased short-interval intracortical and transcallosal inhibition, as well as a normal cortical silent period and long-interval cortical inhibition. These findings were the same during euthymia. Symptoms associated with motor hyperactivity were correlated negatively with the degree of cortical inhibition. These correlations were not significant when a Bonferroni correction was applied. CONCLUSIONS The present longitudinal study showed cortical inhibitory deficits in patients with BD, and supports the hypothesis that cortical inhibitory deficits in BD are trait dependent. Further research is necessary to confirm the clinical significance of these deficits.
Collapse
Affiliation(s)
- Miguel Ruiz-Veguilla
- Grupo Neurodesarrollo y Psicosis, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla/UGC Salud Mental HVR, Seville, Spain
| | - Juan Francisco Martín-Rodríguez
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Francisco J Palomar
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Paolo Porcacchia
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Paloma Álvarez de Toledo
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Salvador Perona-Garcelán
- Grupo Neurodesarrollo y Psicosis, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla/UGC Salud Mental HVR, Seville, Spain
| | - Juan Francisco Rodríguez-Testal
- Grupo Neurodesarrollo y Psicosis, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla/UGC Salud Mental HVR, Seville, Spain.,Departamento de Personalidad, Evaluación y Tratamientos Psicológicos, Facultad de Psicología, Universidad de Sevilla, Seville, Spain
| | - Ismael Huertas-Fernández
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Seville, Spain
| |
Collapse
|
25
|
Kosonsiriluk S, Chaiworakul V, Mauro LJ, El Halawani ME. Enhanced GABAergic inhibition in the premammillary nucleus of photorefractory turkey hens via GABAA receptor upregulation. Gen Comp Endocrinol 2016; 230-231:57-66. [PMID: 27055929 DOI: 10.1016/j.ygcen.2016.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/31/2016] [Accepted: 04/02/2016] [Indexed: 11/20/2022]
Abstract
The premammillary nucleus (PMM) of the turkey mediobasal hypothalamus, where dopamine-melatonin (DA-Mel) neurons are localized, is a site for photoreception and photoperiodic time measurement, which is essential for the initiation of avian reproductive seasonality. In addition, this area could also be responsible for the onset and maintenance of photorefractoriness at the end of the breeding season due to the enhanced inhibitory effect of γ-aminobutyric acid (GABA). GABA is an inhibitory neurotransmitter in the central nervous system which interferes with the photosexual response in the turkey, a seasonally breeding bird. Here, we further characterized the GABAA receptor subunits in the PMM DA-Mel neurons related to reproductive seasonality and the onset of photorefractoriness. GABAA receptor subunits and GABA synthesis enzymes in the PMM of photosensitive and photorefractory turkey hens were identified using real-time qRT-PCR. The upregulation of GABAA receptor α1-3, β2-3, γ1-3, ρ1-3, δ, and θ mRNA expression were observed in the PMM of photorefractory birds when compared to those of photosensitive ones while there is no change observed in the GABA synthesis enzymes, glutamate decarboxylase 1 and 2. Those upregulated GABAA receptor subunits were further examined using immunohistochemical staining and they appeared to be co-localized within the PMM DA-Mel neurons. The upregulation of GABAA receptor subunits observed in the PMM of photorefractory birds coincides with a lack of responsiveness to a light stimulus provided during the photosensitive phase. This is supported by the absence of c-fos induction and TH upregulation in the PMM and a subsequence inhibition of c-fos and GnRH-I expression in the nucleus commissurae pallii. The augmented GABAA receptor subunits expression may mediate an enhancement of inhibitory GABAergic neurotransmission and the subsequent interference with the photosexual response. This could contribute to the state of photorefractoriness and the termination of breeding activities in the turkey, a temperate zone bird.
Collapse
Affiliation(s)
| | - Voravasa Chaiworakul
- Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA
| | - Laura J Mauro
- Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA
| | | |
Collapse
|
26
|
Functional sites involved in modulation of the GABAA receptor channel by the intravenous anesthetics propofol, etomidate and pentobarbital. Neuropharmacology 2016; 105:207-214. [PMID: 26767954 DOI: 10.1016/j.neuropharm.2016.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/15/2015] [Accepted: 01/03/2016] [Indexed: 11/22/2022]
Abstract
GABAA receptors are the major inhibitory neurotransmitter receptors in the brain and are the target for many clinically important drugs. Among the many modulatory compounds are also the intravenous anesthetics propofol and etomidate, and barbiturates. The mechanism of receptor modulation by these compounds is of mayor relevance. The site of action of these compounds has been located to subunit interfaces in the intra-membrane region of the receptor. In α1β2γ2 GABAA receptors there are five such interfaces, two β+/α- and one each of α+/β-, α+/γ- and γ+/β- subunit interfaces. We have used reporter mutations located in the second trans-membrane region in different subunits to probe the effects of changes at these subunit interfaces on modulation by propofol, etomidate and pentobarbital. We provide evidence for the fact that each of these compounds either modulates through a different set of subunit interfaces or through the same set of subunit interfaces to a different degree. As a GABAA receptor pentamer harbors two β+/α- subunit interfaces, we used concatenated receptors to dissect the contribution of individual interfaces and show that only one of these interfaces is important for receptor modulation by etomidate.
Collapse
|
27
|
The Association of the GABRP Polymorphisms with Systemic Lupus Erythematosus. J Immunol Res 2015; 2015:602154. [PMID: 26634217 PMCID: PMC4655060 DOI: 10.1155/2015/602154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/21/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022] Open
Abstract
Gamma-aminobutyric acid receptor subunit pi (GABRP) is involved in inhibitory synaptic transmission in the central nervous system. This gene encodes multisubunit chloride channels and is also expressed in numerous nonneuronal tissues such as the uterus and the ovaries. This study was aimed to validate whether the polymorphisms in the GABRP gene are associated with the susceptibility to systematic lupus erythematosus (SLE). The genotype frequencies of the rs929763, rs732157, and rs3805455 of the GABRP gene in SLE patients were significantly different from those of the control group (P < 0.0001, P = 0.05 and 0.002, resp.). Additional analysis showed that the genotype of the rs929763 and rs3805455 of the GABRP gene were also significantly associated with female SLE patients (P < 0.0001, P = 0.005, resp.). Two haplotype frequencies including a major haplotype of GABRP SNPs were more significantly different between the SLE patients and the healthy controls (P = 0.038 and 4.2E − 24, resp.). These results suggest that the polymorphisms in the GABRP gene might be associated with the susceptibility to SLE and the haplotype of GABRP SNPs is useful genetic marker for SLE.
Collapse
|
28
|
Farb DH, Ratner MH. Targeting the modulation of neural circuitry for the treatment of anxiety disorders. Pharmacol Rev 2015; 66:1002-32. [PMID: 25237115 DOI: 10.1124/pr.114.009126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anxiety disorders are a major public health concern. Here, we examine the familiar area of anxiolysis in the context of a systems-level understanding that will hopefully lead to revealing an underlying pharmacological connectome. The introduction of benzodiazepines nearly half a century ago markedly improved the treatment of anxiety disorders. These agents reduce anxiety rapidly by allosterically enhancing the postsynaptic actions of GABA at inhibitory type A GABA receptors but side effects limit their use in chronic anxiety disorders. Selective serotonin reuptake inhibitors and serotonin/norepinephrine reuptake inhibitors have emerged as an effective first-line alternative treatment of such anxiety disorders. However, many individuals are not responsive and side effects can be limiting. Research into a relatively new class of agents known as neurosteroids has revealed novel modulatory sites and mechanisms of action that are providing insights into the pathophysiology of certain anxiety disorders, potentially bridging the gap between the GABAergic and serotonergic circuits underlying anxiety. However, translating the pharmacological activity of compounds targeted to specific receptor subtypes in rodent models of anxiety to effective therapeutics in human anxiety has not been entirely successful. Since modulating any one of several broad classes of receptor targets can produce anxiolysis, we posit that a systems-level discovery platform combined with an individualized medicine approach based on noninvasive brain imaging would substantially advance the development of more effective therapeutics.
Collapse
Affiliation(s)
- David H Farb
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Marcia H Ratner
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
29
|
Bagci E, Aydin E, Mihasan M, Maniu C, Hritcu L. Anxiolytic and antidepressant-like effects ofFerulago angulataessential oil in the scopolamine rat model of Alzheimer's disease. FLAVOUR FRAG J 2015. [DOI: 10.1002/ffj.3289] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Eyup Bagci
- Department of Biology, Faculty of Science; Firat University; 23119 Elazig Turkey
| | - Emel Aydin
- Department of Biology, Faculty of Science; Firat University; 23119 Elazig Turkey
| | - Marius Mihasan
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| | - Calin Maniu
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| | - Lucian Hritcu
- Department of Biology; Alexandru Ioan Cuza University; Bd. Carol I, No.11 Iasi 700506 Romania
| |
Collapse
|
30
|
Mascarenhas DC, Gomes KS, Nunes-de-Souza RL. Role of TRPV1 channels of the dorsal periaqueductal gray in the modulation of nociception and open elevated plus maze-induced antinociception in mice. Behav Brain Res 2015; 292:547-54. [PMID: 26183651 DOI: 10.1016/j.bbr.2015.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 12/19/2022]
Abstract
Recent findings have identified the presence of transient receptor potential vanilloid-1 (TRPV1) channels within the dorsal portion of the periaqueductal gray (dPAG), suggesting their involvement in the control of pain and environmentally-induced antinociception. Environmentally, antinociception may be achieved through the use of an open elevated plus maze (oEPM, an EPM with 4 open arms), a highly aversive environmental situation. Here, we investigated the role of these TRPV1 channels within the dPAG in the modulation of a tonic pain and in the oEPM-induced antinociception. Male Swiss mice, under the nociceptive effect of 2.5% formalin injected into the right hind paw, received intra-dPAG injections of the TRPV1 agonist (capsaicin: 0, 0.01, 0.1 or 1.0 nmol/0.2 μL; Experiment 1) or antagonist (capsazepine: 0, 10 or 30 nmol/0.2 μL; Experiment 2) or combined injections of capsazepine (30 nmol) and capsaicin (1.0 nmol) (Experiment 3) and the time spent licking the formalin-injected paw was recorded. In Experiment 4, mice received intra-dPAG capsazepine (0 or 30 nmol) and were exposed to the oEPM or to a control situation, an enclosed EPM (eEPM; an EPM with 4 enclosed arms). Results showed that while capsaicin (1 nmol) decreased the time spent licking the formalin-injected paw, capsazepine did not change nociceptive response. Capsazepine (30 nmol) blocked pain inhibition induced by capsaicin and mildly attenuated the oEPM-induced antinociception. Our results revealed an important role of TRPV1 channels within the dPAG in the modulation of pain and in the phenomenon known as fear-induced antinociception in mice.
Collapse
Affiliation(s)
- Diego Cardozo Mascarenhas
- Joint Graduate Program in Physiological Sciences, UFSCar/UNESP-São Carlos, SP, 13565-905, Brazil; School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, 14801-902, Araraquara, SP, Brazil
| | - Karina Santos Gomes
- School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, 14801-902, Araraquara, SP, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences, UFSCar/UNESP-São Carlos, SP, 13565-905, Brazil; School of Pharmaceutical Sciences, Universidade Estadual Paulista-UNESP, 14801-902, Araraquara, SP, Brazil.
| |
Collapse
|
31
|
Wong LW, Tae HS, Cromer BA. Assembly, trafficking and function of α1β2γ2 GABAA receptors are regulated by N-terminal regions, in a subunit-specific manner. J Neurochem 2015; 134:819-32. [PMID: 26016529 DOI: 10.1111/jnc.13175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/16/2015] [Accepted: 05/09/2015] [Indexed: 02/01/2023]
Abstract
GABAA receptors are pentameric ligand-gated ion channels that mediate inhibitory fast synaptic transmission in the central nervous system. Consistent with recent pentameric ligand-gated ion channels structures, sequence analysis predicts an α-helix near the N-terminus of each GABAA receptor subunit. Preceding each α-helix are 8-36 additional residues, which we term the N-terminal extension. In homomeric GABAC receptors and nicotinic acetylcholine receptors, the N-terminal α-helix is functionally essential. Here, we determined the role of the N-terminal extension and putative α-helix in heteromeric α1β2γ2 GABAA receptors. This role was most prominent in the α1 subunit, with deletion of the N-terminal extension or further deletion of the putative α-helix both dramatically reduced the number of functional receptors at the cell surface. Conversely, deletion of the β2 or γ2 N-terminal extension had little effect on the number of functional cell surface receptors. Additional deletion of the putative α-helix in the β2 or γ2 subunits did, however, decrease both functional cell surface receptors and incorporation of the γ2 subunit into mature receptors. In the β2 subunit only, α-helix deletions affected GABA sensitivity and desensitization. Our findings demonstrate that N-terminal extensions and α-helices make key subunit-specific contributions to assembly, consistent with both regions being involved in inter-subunit interactions. N-terminal α-helices and preceding sequences of eukaryotic pentameric ligand-gated ion channels are absent in prokaryotic homologues, suggesting they may not be functionally essential. Here, we show that in heteropentameric α1β2γ2 GABAA receptors, the role of these segments is highly subunit dependent. The extension preceding the α-helix in the α subunit is crucial for assembly and trafficking, but is of little importance in β and γ subunits. Indeed, robust receptor levels remain when the extension and α-helix are removed in β or γ subunits.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Vic., Australia
| | - Han-Shen Tae
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Brett A Cromer
- Health Innovation Research Institute, School of Medical Sciences, RMIT University, Melbourne, Vic., Australia
| |
Collapse
|
32
|
Jutfelt F, Hedgärde M. Juvenile Atlantic cod behavior appears robust to near-future CO2 levels. Front Zool 2015; 12:11. [PMID: 27408612 PMCID: PMC4940919 DOI: 10.1186/s12983-015-0104-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/29/2015] [Indexed: 12/01/2022] Open
Abstract
Background Ocean acidification caused by the anthropogenic release of CO2 is considered a major threat to marine ecosystems. One unexpected impact of elevated water CO2 levels is that behavioral alterations may occur in tropical reef fish and certain temperate fish species. These effects appear to alter many different types of sensory and cognitive functions; if widespread and persistent, they have the potential to cause ecosystem changes. Methods We investigated whether economically and ecologically important Atlantic cod also display behavioral abnormalities by exposing 52 juvenile cod to control conditions (500 μatm, duplicate tanks) or an end-of-the-century ocean acidification scenario (1000 μatm, duplicate tanks) for one month, during which time the fish were examined for a range of behaviors that have been reported to be affected by elevated CO2 in other fish. The behaviors were swimming activity, as measured by number of lines crossed per minute, the emergence from shelter, determined by how long it took the fish to exit a shelter after a disturbance, relative lateralization (a measure of behavioral turning side preference), and absolute lateralization (the strength of behavioral symmetry). Results We found no effect of CO2 treatment on any of the four behaviors tested: activity (F = 1.61, p = 0.33), emergence from shelter (F = 0.13, p = 0.76), relative lateralization (F = 2.82, p = 0.50), and absolute lateralization (F = 0.80, p = 0.26). Conclusion Our results indicate that the behavior of Atlantic cod could be resilient to the impacts of near-future levels of water CO2.
Collapse
Affiliation(s)
- Fredrik Jutfelt
- Department of Biological and Environmental Sciences, University of Gothenburg, PO Box 463, SE-405 30 Göteborg, Sweden ; The Lovén Centre Kristineberg, Kristineberg 566, SE-451 78 Fiskebäckskil, Sweden
| | - Maria Hedgärde
- Department of Biological and Environmental Sciences, University of Gothenburg, PO Box 463, SE-405 30 Göteborg, Sweden
| |
Collapse
|
33
|
Hénin J, Salari R, Murlidaran S, Brannigan G. A predicted binding site for cholesterol on the GABAA receptor. Biophys J 2014; 106:1938-49. [PMID: 24806926 DOI: 10.1016/j.bpj.2014.03.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/29/2022] Open
Abstract
Modulation of the GABA type A receptor (GABAAR) function by cholesterol and other steroids is documented at the functional level, yet its structural basis is largely unknown. Current data on structurally related modulators suggest that cholesterol binds to subunit interfaces between transmembrane domains of the GABAAR. We construct homology models of a human GABAAR based on the structure of the glutamate-gated chloride channel GluCl of Caenorhabditis elegans. The models show the possibility of previously unreported disulfide bridges linking the M1 and M3 transmembrane helices in the α and γ subunits. We discuss the biological relevance of such disulfide bridges. Using our models, we investigate cholesterol binding to intersubunit cavities of the GABAAR transmembrane domain. We find that very similar binding modes are predicted independently by three approaches: analogy with ivermectin in the GluCl crystal structure, automated docking by AutoDock, and spontaneous rebinding events in unbiased molecular dynamics simulations. Taken together, the models and atomistic simulations suggest a somewhat flexible binding mode, with several possible orientations. Finally, we explore the possibility that cholesterol promotes pore opening through a wedge mechanism.
Collapse
Affiliation(s)
- Jérôme Hénin
- Laboratoire de Biochimie Théorique, CNRS, IBPC, and Université Paris Diderot, Paris, France
| | - Reza Salari
- Department of Physics, Rutgers University-Camden, Camden, New Jersey; Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, New Jersey
| | - Sruthi Murlidaran
- Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, New Jersey
| | - Grace Brannigan
- Department of Physics, Rutgers University-Camden, Camden, New Jersey; Center for Computational and Integrative Biology, Rutgers University-Camden, Camden, New Jersey.
| |
Collapse
|
34
|
Middendorp SJ, Hurni E, Schönberger M, Stein M, Pangerl M, Trauner D, Sigel E. Relative positioning of classical benzodiazepines to the γ2-subunit of GABAA receptors. ACS Chem Biol 2014; 9:1846-53. [PMID: 24918742 DOI: 10.1021/cb500186a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
GABAA receptors are the major inhibitory neurotransmitter receptors in the brain. Benzodiazepine exert their action via a high affinity-binding site at the α/γ subunit interface on some of these receptors. Diazepam has sedative, hypnotic, anxiolytic, muscle relaxant, and anticonvulsant effects. It acts by potentiating the current evoked by the agonist GABA. Understanding specific interaction of benzodiazepines in the binding pocket of different GABAA receptor isoforms might help to separate these divergent effects. As a first step, we characterized the interaction between diazepam and the major GABAA receptor isoform α1β2γ2. We mutated several amino acid residues on the γ2-subunit assumed to be located near or in the benzodiazepine binding pocket individually to cysteine and studied the interaction with three ligands that are modified with a cysteine-reactive isothiocyanate group (-NCS). When the reactive NCS group is in apposition to the cysteine residue this leads to a covalent reaction. In this way, three amino acid residues, γ2Tyr58, γ2Asn60, and γ2Val190 were located relative to classical benzodiazepines in their binding pocket on GABAA receptors.
Collapse
Affiliation(s)
- Simon J. Middendorp
- Institute
of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Evelyn Hurni
- Institute
of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| | - Matthias Schönberger
- Department
of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, 81377 Munich, Germany
| | - Marco Stein
- Department
of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, 81377 Munich, Germany
| | - Michael Pangerl
- Department
of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, 81377 Munich, Germany
| | - Dirk Trauner
- Department
of Chemistry, Ludwig-Maximilians-Universität München and Center of Integrated Protein Science, 81377 Munich, Germany
| | - Erwin Sigel
- Institute
of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland
| |
Collapse
|
35
|
Baur R, Schuehly W, Sigel E. Moderate concentrations of 4-O-methylhonokiol potentiate GABAA receptor currents stronger than honokiol. Biochim Biophys Acta Gen Subj 2014; 1840:3017-21. [PMID: 24973566 DOI: 10.1016/j.bbagen.2014.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Magnolia bark preparations from Magnolia officinalis of Asian medicinal systems are known for their muscle relaxant effect and anticonvulsant activity. These CNS related effects are ascribed to the presence of the biphenyl-type neolignans honokiol and magnolol that exert a potentiating effect on GABAA receptors. 4-O-methylhonokiol isolated from seeds of the North-American M. grandiflora was compared to honokiol for its activity to potentiate GABAA receptors and its GABAA receptor subtype-specificity was established. METHODS Different recombinant GABAA receptors were functionally expressed in Xenopus oocytes and electrophysiological techniques were used determine to their modulation by 4-O-methylhonokiol. RESULTS 3μM 4-O-methylhonokiol is shown here to potentiate responses of the α₁β₂γ₂ GABAA receptor about 20-fold stronger than the same concentration of honokiol. In the present study potentiation by 4-O-methylhonokiol is also detailed for 12 GABAA receptor subtypes to assess GABAA receptor subunits that are responsible for the potentiating effect. CONCLUSION The much higher potentiation of GABAA receptors at identical concentrations of 4-O-methylhonokiol as compared to honokiol parallels previous observations made in other systems of potentiated pharmacological activity of 4-O-methylhonokiol over honokiol. GENERAL SIGNIFICANCE The results point to the use of 4-O-methylhonokiol as a lead for GABAA receptor potentiation and corroborate the use of M. grandiflora seeds against convulsions in Mexican folk medicine.
Collapse
Affiliation(s)
- Roland Baur
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstr. 28, CH-3012 Bern, Switzerland
| | - Wolfgang Schuehly
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy, Karl-Franzens-University, Universitätsplatz 4, 8010 Graz, Austria.
| | - Erwin Sigel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstr. 28, CH-3012 Bern, Switzerland
| |
Collapse
|
36
|
Abstract
Premenstrual dysphoric disorder (PMDD) is comprised of a cluster of affective, behavioral and somatic symptoms recurring monthly during the luteal phase of the menstrual cycle. The disorder affects 3-8% of menstruating women and represents the more severe and disabling end of the spectrum of premenstrual disorders, which includes premenstrual syndrome and premenstrual aggravation of underlying affective disorder. Rigorous and specific diagnostic criteria for PMDD were specified in the Diagnostic and Statistical Manual of Mental Disorders IV (1994) and reaffirmed in the Diagnostic and Statistical Manual of Mental Disorders V (2013) and, consequently, there has been a marked increase in well-designed, placebo-controlled studies evaluating treatment modalities. Although the exact pathogenesis of PMDD is still elusive, treatment of PMDD and severe premenstrual syndrome has centered on neuromodulation via serotonin reuptake inhibitor antidepressants, and ovulation suppression utilizing various contraceptive and hormonal preparations. Unlike the approach to the treatment of depression, serotonergic antidepressants need not be given daily, but can be effective when used cyclically, only in the luteal phase or even limited to the duration of the monthly symptoms. Less, well-substantiated alternative treatments, such as calcium supplementation, agnus castus (chasteberry), Hypericum perforatum (St John's wort) and cognitive/behavioral/relaxation therapies, may be useful adjuncts in the treatment of PMDD. This review provides an overview of current information on the treatment of PMDD.
Collapse
Affiliation(s)
- Andrea J Rapkin
- Department of Obstetrics & Gynecology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 10833 Le Conte Avenue, Room 27-139 CHS, Los Angeles, CA 90095-1740, USA
| | | |
Collapse
|
37
|
Miller PS, Aricescu AR. Crystal structure of a human GABAA receptor. Nature 2014; 512:270-5. [PMID: 24909990 PMCID: PMC4167603 DOI: 10.1038/nature13293] [Citation(s) in RCA: 547] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 03/28/2014] [Indexed: 01/01/2023]
Abstract
Type-A γ-aminobutyric acid receptors (GABAARs) are the principal mediators of rapid inhibitory synaptic transmission in the human brain. A decline in GABAAR signalling triggers hyperactive neurological disorders such as insomnia, anxiety and epilepsy. Here we present the first three-dimensional structure of a GABAAR, the human β3 homopentamer, at 3 Å resolution. This structure reveals architectural elements unique to eukaryotic Cys-loop receptors, explains the mechanistic consequences of multiple human disease mutations and shows an unexpected structural role for a conserved N-linked glycan. The receptor was crystallized bound to a previously unknown agonist, benzamidine, opening a new avenue for the rational design of GABAAR modulators. The channel region forms a closed gate at the base of the pore, representative of a desensitized state. These results offer new insights into the signalling mechanisms of pentameric ligand-gated ion channels and enhance current understanding of GABAergic neurotransmission.
Collapse
Affiliation(s)
- Paul S Miller
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
38
|
Thore SN, Gupta SV, Baheti KG. Synthesis and Pharmacological Evaluation of Novel Triazolo [4, 3-a] tetrahydrobenzo (b) thieno [3, 2-e] pyrimidine-5(4H)-ones. J Heterocycl Chem 2014. [DOI: 10.1002/jhet.1989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- S. N. Thore
- Deogiri College; Padampura Aurangabad 431005 India
| | | | | |
Collapse
|
39
|
Martínez-Cué C, Delatour B, Potier MC. Treating enhanced GABAergic inhibition in Down syndrome: use of GABA α5-selective inverse agonists. Neurosci Biobehav Rev 2014; 46 Pt 2:218-27. [PMID: 24412222 DOI: 10.1016/j.neubiorev.2013.12.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/27/2013] [Accepted: 12/16/2013] [Indexed: 11/27/2022]
Abstract
Excess inhibition in the brain of individuals carrying an extra copy of chromosome 21 could be responsible for cognitive deficits observed throughout their lives. A change in the excitatory/inhibitory balance in adulthood would alter synaptic plasticity, potentially triggering learning and memory deficits. γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the mature central nervous system and binds to GABAA receptors, opens a chloride channel, and reduces neuronal excitability. In this review we discuss methods to alleviate neuronal inhibition in a mouse model of Down syndrome, the Ts65Dn mouse, using either an antagonist (pentylenetetrazol) or two different inverse agonists selective for the α5-subunit containing receptor. Both inverse agonists, which reduce inhibitory GABAergic transmission, could rescue learning and memory deficits in Ts65Dn mice. We also discuss safety issues since modulation of the excitatory-inhibitory balance to improve cognition without inducing seizures remains particularly difficult when using GABA antagonists.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Benoît Delatour
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau et de Moelle Epinière, CNRS UMR7225, INSERM U1127, UPMC, IHUA-ICM, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
40
|
Hashimoto T, Kiyoshi T, Kohayakawa H, Iwamura Y, Yoshida N. Pharmacological properties of AC-3933, a novel benzodiazepine receptor partial inverse agonist. Neuroscience 2014; 256:352-9. [DOI: 10.1016/j.neuroscience.2013.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
41
|
Klepeisz P, Sagmeister S, Haudek-Prinz V, Pichlbauer M, Grasl-Kraupp B, Gerner C. Phenobarbital induces alterations in the proteome of hepatocytes and mesenchymal cells of rat livers. PLoS One 2013; 8:e76137. [PMID: 24204595 PMCID: PMC3812042 DOI: 10.1371/journal.pone.0076137] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 08/27/2013] [Indexed: 11/18/2022] Open
Abstract
Preceding studies on the mode of action of non-genotoxic hepatocarcinogens (NGCs) have concentrated on alterations induced in hepatocytes (HCs). A potential role of non-parenchymal liver cells (NPCs) in NGC-driven hepatocarcinogenesis has been largely neglected so far. The aim of this study is to characterize NGC-induced alterations in the proteome profiles of HCs as well as NPCs. We chose the prototypic NGC phenobarbital (PB) which was applied to male rats for a period of 14 days. The livers of PB-treated rats were perfused by collagenase and the cell suspensions obtained were subjected to density gradient centrifugation to separate HCs from NPCs. In addition, HCs and NPC isolated from untreated animals were treated with PB in vitro. Proteome profiling was done by CHIP-HPLC and ion trap mass spectrometry. Proteome analyses of the in vivo experiments showed many of the PB effects previously described in HCs by other methods, e.g. induction of phase I and phase II drug metabolising enzymes. In NPCs proteins related to inflammation and immune regulation such as PAI-1 and S100-A10, ADP-ribosyl cyclase 1 and to cell migration such as kinesin-1 heavy chain, myosin regulatory light chain RLC-A and dihydropyrimidinase-related protein 1 were found to be induced, indicating major PB effects on these cells. Remarkably, in vitro treatment of HCs and NPCs with PB hardly reproduced the proteome alterations observed in vivo, indicating differences of NGC induced responses of cells at culture conditions compared to the intact organism. To conclude, the present study clearly demonstrated that PB induces proteome alterations not only in HCs but also in NPCs. Thus, any profound molecular understanding on the mode of action of NGCs has to consider effects on cells of the hepatic mesenchyme.
Collapse
Affiliation(s)
- Philip Klepeisz
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Sandra Sagmeister
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Verena Haudek-Prinz
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Melanie Pichlbauer
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Bettina Grasl-Kraupp
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Inner Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
42
|
Saha S, Hu Y, Martin SC, Bandyopadhyay S, Russek SJ, Farb DH. Polycomblike protein PHF1b: a transcriptional sensor for GABA receptor activity. BMC Pharmacol Toxicol 2013; 14:37. [PMID: 23879974 PMCID: PMC3734045 DOI: 10.1186/2050-6511-14-37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/17/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The γ-aminobutyric acid (GABA) type A receptor (GABA(A)R) contains the recognition sites for a variety of agents used in the treatment of brain disorders, including anxiety and epilepsy. A better understanding of how receptor expression is regulated in individual neurons may provide novel opportunities for therapeutic intervention. Towards this goal we have studied transcription of a GABA(A)R subunit gene (GABRB1) whose activity is autologously regulated by GABA via a 10 base pair initiator-like element (β(1)-INR). METHODS By screening a human cDNA brain library with a yeast one-hybrid assay, the Polycomblike (PCL) gene product PHD finger protein transcript b (PHF1b) was identified as a β(1)-INR associated protein. Promoter/reporter assays in primary rat cortical cells demonstrate that PHF1b is an activator at GABRB1, and chromatin immunoprecipitation assays reveal that presence of PHF1 at endogenous Gabrb1 is regulated by GABA(A)R activation. RESULTS PCL is a member of the Polycomb group required for correct spatial expression of homeotic genes in Drosophila. We now show that PHF1b recognition of β(1)-INR is dependent on a plant homeodomain, an adjacent helix-loop-helix, and short glycine rich motif. In neurons, it co-immunoprecipitates with SUZ12, a key component of the Polycomb Repressive Complex 2 (PRC2) that regulates a number of important cellular processes, including gene silencing via histone H3 lysine 27 trimethylation (H3K27me3). CONCLUSIONS The observation that chronic exposure to GABA reduces PHF1 binding and H3K27 monomethylation, which is associated with transcriptional activation, strongly suggests that PHF1b may be a molecular transducer of GABA(A)R function and thus GABA-mediated neurotransmission in the central nervous system.
Collapse
Affiliation(s)
- Shamol Saha
- Department of Pharmacology & Experimental Therapeutics, Laboratory of Translational Epilepsy, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | |
Collapse
|
43
|
Kurata K, Nagasawa M, Tomonaga S, Aoki M, Morishita K, Denbow DM, Furuse M. Orally administeredl-ornithine elevates brainl-ornithine levels and has an anxiolytic-like effect in mice. Nutr Neurosci 2013; 14:243-8. [DOI: 10.1179/1476830511y.0000000018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
44
|
Anxiolytic effects of Julibroside C1 isolated from Albizzia julibrissin in mice. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:184-92. [PMID: 23481219 DOI: 10.1016/j.pnpbp.2013.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 02/08/2013] [Accepted: 02/21/2013] [Indexed: 11/24/2022]
Abstract
Julibroside C1 is a saponin-containing compound isolated from Albizzia julibrissin Durazz. In this study, we investigated the putative anxiolytic effects of Julibroside C1 using the elevated plus maze (EPM) in mice. Julibroside C1 at doses of 0.5 and 1 mg/kg significantly increased the time spent in the open arms and the number of entries into the open arms of the EPM compared to the control group. Moreover, the anxiolytic-like effects of Julibroside C1 (0.5 mg/kg) were blocked by WAY-100635 (5-HT1A receptor antagonist), bicuculline (GABA(A) receptor antagonist), and flumazenil (antagonist of the GABA(A) receptor benzodiazepine site). However, Julibroside C1 did not change locomotor activity or induce myorelaxant effects. We used quantitative receptor autoradiography to investigate the effects of Julibroside C1 on alterations in mouse brain receptors. After acute treatment with Julibroside C1 (0.5 mg/kg), [(3)H]-8-OH-DPAT binding was significantly decreased in the CA1 region of the hippocampus and [(3)H]-flunitrazepam binding was decreased remarkably in the cingulate cortex region. However, [(3)H]-muscimol binding did not show a significant change in any brain region. Taken together, our findings suggest that Julibroside C1 shows anxiolytic-like effects, which might be mediated by the 5-HT1A and GABA(A)-benzodiazepine receptor systems.
Collapse
|
45
|
Behavioural disturbances in a temperate fish exposed to sustained high-CO2 levels. PLoS One 2013; 8:e65825. [PMID: 23750274 PMCID: PMC3672104 DOI: 10.1371/journal.pone.0065825] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/30/2013] [Indexed: 01/18/2023] Open
Abstract
As atmospheric CO2 levels rise, the CO2 concentration in ocean surface waters increases through a process commonly referred to as ocean acidification. Recently, surprising behavioural modifications has been detected in the early life stages of tropical coral reef fish exposed to ocean acidification-relevant CO2 concentrations, but it has been unclear if this effect could occur in temperate waters. Here we show several severe behavioural disturbances, including effects on boldness, exploratory behaviour, lateralisation, and learning in a temperate fish, the three-spined stickleback (Gasterosteus aculeatus). The behavioural effects were consistent throughout the exposure period and increased in effect size with exposure time. We observed the effects on adult sticklebacks, a species known to be tolerant to other environmental stressors. Our findings suggest that behavioural abnormalities that stem from CO2 exposure are not restricted to sensitive tropical species or early life stages and may therefore affect fish on a global scale. The severity of disturbances and the possibility of a serious behavioural problem for fish across the globe is cause for concern.
Collapse
|
46
|
Caspary DM, Hughes LF, Ling LL. Age-related GABAA receptor changes in rat auditory cortex. Neurobiol Aging 2012; 34:1486-96. [PMID: 23257264 DOI: 10.1016/j.neurobiolaging.2012.11.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 11/13/2012] [Accepted: 11/16/2012] [Indexed: 01/06/2023]
Abstract
Auditory cortex (AI) shows age-related decreases in pre-synaptic markers for gamma-aminobutyric acid (GABA) and degraded AI neuronal response properties. Previous studies find age-related increases in spontaneous and driven activity, decreased spectral and directional sensitivity, and impaired novelty detection. The present study examined expression of GABA(A) receptor (GABA(A)R) subunit message, protein, and quantitative GABA(A)R binding in young, middle-aged, and aged rat AI, with comparisons with adjoining parietal cortex. Significant loss of GABA(A)R α(1) subunit message across AI layers was observed in middle-aged and aged rats and α(1) subunit protein levels declined in layers II and III. Age-related increases in GABA(A)R α(3) subunit message and protein levels were observed in certain AI layers. GABA(A)R subunits, including β(1), β(2), γ(1), γ(2s), and γ(2L), primarily, but not exclusively, showed age-related declines at the message and protein levels. The ability of GABA to modulate [(3)H]t-butylbicycloorthobenzoate binding in the chloride channel showed age-related decreases in peak binding and changes in desensitization kinetics. Collectively, age-related changes in GABA(A)R subunit composition would alter the magnitude and temporal properties of inhibitory synaptic transmission and could underpin observed age-related functional changes seen in the elderly.
Collapse
Affiliation(s)
- Donald M Caspary
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | | | | |
Collapse
|
47
|
Gale C, Herbison GP, Glue P, Coverdale J, Guaiana G. Benzodiazepines for generalised anxiety disorder (GAD). Cochrane Database Syst Rev 2012. [DOI: 10.1002/14651858.cd001846.pub3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Christopher Gale
- University of Otago; Department of Psychological Medicine; Dunedin School of Medicine PO Box 913 Dunedin New Zealand 9010
| | - G Peter Herbison
- Dunedin School of Medicine, University of Otago; Department of Preventive & Social Medicine; PO Box 913 Dunedin New Zealand 9054
| | - Paul Glue
- University of Otago; Department of Psychological Medicine; Dunedin School of Medicine PO Box 913 Dunedin New Zealand 9010
| | - John Coverdale
- Baylor College of Medicine, Texas Medical Centre; Menninger Department of Psychiatry; Houston Texas USA
| | - Giuseppe Guaiana
- University of Western Ontario; Department of Psychiatry; Regional Mental Health Care-Saint Thomas 467 Sunset Drive St Thomas Ontario Canada N6P 3V9
| |
Collapse
|
48
|
Abstract
The GABA(A) receptors are the major inhibitory neurotransmitter receptors in mammalian brain. Each isoform consists of five homologous or identical subunits surrounding a central chloride ion-selective channel gated by GABA. How many isoforms of the receptor exist is far from clear. GABA(A) receptors located in the postsynaptic membrane mediate neuronal inhibition that occurs in the millisecond time range; those located in the extrasynaptic membrane respond to ambient GABA and confer long-term inhibition. GABA(A) receptors are responsive to a wide variety of drugs, e.g. benzodiazepines, which are often used for their sedative/hypnotic and anxiolytic effects.
Collapse
Affiliation(s)
- Erwin Sigel
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012 Bern, Switzerland.
| | | |
Collapse
|
49
|
Lüscher BP, Baur R, Goeldner M, Sigel E. Influence of GABA(A) receptor α subunit isoforms on the benzodiazepine binding site. PLoS One 2012; 7:e42101. [PMID: 22848717 PMCID: PMC3407089 DOI: 10.1371/journal.pone.0042101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/02/2012] [Indexed: 11/20/2022] Open
Abstract
Classical benzodiazepines, such as diazepam, interact with αxβ2γ2 GABAA receptors, x = 1, 2, 3, 5 and modulate their function. Modulation of different receptor isoforms probably results in selective behavioural effects as sedation and anxiolysis. Knowledge of differences in the structure of the binding pocket in different receptor isoforms is of interest for the generation of isoform-specific ligands. We studied here the interaction of the covalently reacting diazepam analogue 3-NCS with α1S204Cβ2γ2, α1S205Cβ2γ2 and α1T206Cβ2γ2 and with receptors containing the homologous mutations in α2β2γ2, α3β2γ2, α5β1/2γ2 and α6β2γ2. The interaction was studied using radioactive ligand binding and at the functional level using electrophysiological techniques. Both strategies gave overlapping results. Our data allow conclusions about the relative apposition of α1S204Cβ2γ2, α1S205Cβ2γ2 and α1T206Cβ2γ2 and homologous positions in α2, α3, α5 and α6 with C-atom adjacent to the keto-group in diazepam. Together with similar data on the C-atom carrying Cl in diazepam, they indicate that the architecture of the binding site for benzodiazepines differs in each GABAA receptor isoform α1β2γ2, α2β2γ2, α3β2γ2, α5β1/2γ2 and α6β2γ2.
Collapse
Affiliation(s)
- Benjamin P. Lüscher
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Roland Baur
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Maurice Goeldner
- Laboratoire de Conception et Application de Molécules Bioactives, Unité Mixte de Recherche CNRS, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Erwin Sigel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Baur R, Gertsch J, Sigel E. The cannabinoid CB1 receptor antagonists rimonabant (SR141716) and AM251 directly potentiate GABA(A) receptors. Br J Pharmacol 2012; 165:2479-84. [PMID: 21470203 DOI: 10.1111/j.1476-5381.2011.01405.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Rimonabant (SR141716) and the structurally related AM251 are widely used in pharmacological experiments as selective cannabinoid receptor CB(1) antagonists / inverse agonists. Concentrations of 0.5-10 µM are usually applied in in vitro experiments. We intended to show that these drugs did not act at GABA(A) receptors but found a significant positive allosteric modulation instead. EXPERIMENTAL APPROACH Recombinant GABA(A) receptors were expressed in Xenopus oocytes. Receptors were exposed to AM251 or rimonabant in the absence and presence of GABA. Standard electrophysiological techniques were used to monitor the elicited ionic currents. KEY RESULTS AM251 dose-dependently potentiated responses to 0.5 µM GABA at the recombinant α(1) β(2) γ(2) GABA(A) receptor with an EC(50) below 1 µM and a maximal potentiation of about eightfold. The Hill coefficient indicated that more than one binding site for AM251 was located in this receptor. Rimonabant had a lower affinity, but a fourfold higher efficacy. AM251 potentiated also currents mediated by α(1) β(2) , α(x) β(2) γ(2) (x = 2,3,5,6), α(1) β(3) γ(2) and α(4) β(2) δ GABA(A) receptors, but not those mediated by α(1) β(1) γ(2) . Interestingly, the CB(1) receptor antagonists LY320135 and O-2050 did not significantly affect α(1) β(2) γ(2) GABA(A) receptor-mediated currents at concentrations of 1 µM. CONCLUSIONS AND IMPLICATIONS This study identified rimonabant and AM251 as positive allosteric modulators of GABA(A) receptors. Thus, potential GABAergic effects of commonly used concentrations of these compounds should be considered in in vitro experiments, especially at extrasynaptic sites where GABA concentrations are low. LINKED ARTICLES This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.
Collapse
Affiliation(s)
- R Baur
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstr, Bern, Switzerland
| | | | | |
Collapse
|