51
|
Maltsev DV, Spasov AA, Yakovlev DS, Vassiliev PM, Skripka MO, Miroshnikov MV, Sultanova KT, Kochetkov AN, Divaeva LN, Kuzmenko TA, Morkovnik AS. Searching for new anxiolytic agents among derivatives of 11-dialkylaminoethyl-2,3,4,5-tetrahydrodiazepino[1,2-a]benzimidazole. Eur J Pharm Sci 2021; 161:105792. [PMID: 33705965 DOI: 10.1016/j.ejps.2021.105792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
A study on the anxiolytic activity of the new derivatives of 11-dialkylaminoethyl-2,3,4,5-tetrahydrodiazepino[1,2-a]benzimidazole, containing privileged scaffolds of benzodiazepine and benzimidazole in their structure, was conducted. The cytotoxic properties of low levels of six compounds were preliminary determined in vitro using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide test. The screening of these substances for anxiolytic activity was conducted using elevated plus maze (EPM) test in vivo, and DAB-21 was found to be the most active compound. The acute toxicity of DAB-21 was determined as less toxic than that of diazepam. The dose-dependent effect of the most active compound revealed a minimum dose of 1.26 mg/kg, which resulted in the maximum counterphobic effect. The effect of DAB-21 was superior in a number of tests compared with that of diazepam, which indicated a high level of tranquilizing activity for DAB-21. The results of in silico docking analysis suggest that DAB-21 should have a slightly lower anxiolytic activity than diazepam, but should exhibit greater specific affinity for the benzodiazepine site of the GABAA receptor, in comparison with its GABA-binding site. The interaction between DAB-21 and flumazenil in terms of EPM verifies the GABAergic mechanism of action of DAB-21. Our results highlight the potential of 11-dialkylaminomethyl-2,3,4,5-tetrahydrodiazepino[1,2-a]benzimidazoles as promising compounds in the search for new highly effective anxiolytics.
Collapse
Affiliation(s)
- Dmitriy V Maltsev
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia; Volgograd Medical Research Center; 1 Pavshikh Bortsov sq., Volgograd, Russia.
| | - Alexander A Spasov
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia; Volgograd Medical Research Center; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Dmitriy S Yakovlev
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia; Volgograd Medical Research Center; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Pavel M Vassiliev
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Maria O Skripka
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Mikhail V Miroshnikov
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Kira T Sultanova
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia; Volgograd Medical Research Center; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Andrey N Kochetkov
- Volgograd State Medical University, Department of Pharmacology and Bioinformatics; 1 Pavshikh Bortsov sq., Volgograd, Russia
| | - Lyudmila N Divaeva
- Southern Federal University, Research Institute of Physical and Organic Chemistry; 105/42 Bolshaya Sadovaya str., Rostov-on-Don, Russia
| | - Tatyana A Kuzmenko
- Southern Federal University, Research Institute of Physical and Organic Chemistry; 105/42 Bolshaya Sadovaya str., Rostov-on-Don, Russia
| | - Anatolii S Morkovnik
- Southern Federal University, Research Institute of Physical and Organic Chemistry; 105/42 Bolshaya Sadovaya str., Rostov-on-Don, Russia
| |
Collapse
|
52
|
Peters KZ, Cheer JF, Tonini R. Modulating the Neuromodulators: Dopamine, Serotonin, and the Endocannabinoid System. Trends Neurosci 2021; 44:464-477. [PMID: 33674134 DOI: 10.1016/j.tins.2021.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/04/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022]
Abstract
Dopamine (DA), serotonin (5-hydroxytryptamine, 5-HT), and endocannabinoids (ECs) are key neuromodulators involved in many aspects of motivated behavior, including reward processing, reinforcement learning, and behavioral flexibility. Among the longstanding views about possible relationships between these neuromodulators is the idea of DA and 5-HT acting as opponents. This view has been challenged by emerging evidence that 5-HT supports reward seeking via activation of DA neurons in the ventral tegmental area. Adding an extra layer of complexity to these interactions, the endocannabinoid system is uniquely placed to influence dopaminergic and serotonergic neurotransmission. In this review we discuss how these three neuromodulatory systems interact at the cellular and circuit levels. Technological advances that facilitate precise identification and control of genetically targeted neuronal populations will help to achieve a better understanding of the complex relationship between these essential systems, and the potential relevance for motivated behavior.
Collapse
Affiliation(s)
- Kate Z Peters
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD, USA.
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Fondazione Istituto Italiano di Tecnologia, via Morego 30, Genova, Italy.
| |
Collapse
|
53
|
The Distinct Functions of Dopaminergic Receptors on Pain Modulation: A Narrative Review. Neural Plast 2021; 2021:6682275. [PMID: 33688340 PMCID: PMC7920737 DOI: 10.1155/2021/6682275] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/06/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic pain is considered an economic burden on society as it often results in disability, job loss, and early retirement. Opioids are the most common analgesics prescribed for the management of moderate to severe pain. However, chronic exposure to these drugs can result in opioid tolerance and opioid-induced hyperalgesia. On pain modulation strategies, exploiting the multitarget drugs with the ability of the superadditive or synergistic interactions attracts more attention. In the present report, we have reviewed the analgesic effects of different dopamine receptors, particularly D1 and D2 receptors, in different regions of the central nervous system, including the spinal cord, striatum, nucleus accumbens (NAc), and periaqueductal gray (PAG). According to the evidence, these regions are not only involved in pain modulation but also express a high density of DA receptors. The findings can be categorized as follows: (1) D2-like receptors may exert a higher analgesic potency, but D1-like receptors act in different manners across several mechanisms in the mentioned regions; (2) in the spinal cord and striatum, antinociception of DA is mainly mediated by D2-like receptors, while in the NAc and PAG, both D1- and D2-like receptors are involved as analgesic targets; and (3) D2-like receptor agonists can act as adjuvants of μ-opioid receptor agonists to potentiate analgesic effects and provide a better approach to pain relief.
Collapse
|
54
|
Yin JB, Liang SH, Li F, Zhao WJ, Bai Y, Sun Y, Wu ZY, Ding T, Sun Y, Liu HX, Lu YC, Zhang T, Huang J, Chen T, Li H, Chen ZF, Cao J, Ren R, Peng YN, Yang J, Zang WD, Li X, Dong YL, Li YQ. dmPFC-vlPAG projection neurons contribute to pain threshold maintenance and antianxiety behaviors. J Clin Invest 2021; 130:6555-6570. [PMID: 32841213 DOI: 10.1172/jci127607] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
The dorsal medial prefrontal cortex (dmPFC) has been recognized as a key cortical area for nociceptive modulation. However, the underlying neural pathway and the function of specific cell types remain largely unclear. Here, we show that lesions in the dmPFC induced an algesic and anxious state. Using multiple tracing methods including a rabies-based transsynaptic tracing method, we outlined an excitatory descending neural pathway from the dmPFC to the ventrolateral periaqueductal gray (vlPAG). Specific activation of the dmPFC/vlPAG neural pathway by optogenetic manipulation produced analgesic and antianxiety effects in a mouse model of chronic pain. Inhibitory neurons in the dmPFC were specifically activated using a chemogenetic approach, which logically produced an algesic and anxious state under both normal and chronic pain conditions. Antagonists of the GABAA receptor (GABAAR) or mGluR1 were applied to the dmPFC, which produced analgesic and antianxiety effects. In summary, the results of our study suggest that the dmPFC/vlPAG neural pathway might participate in the maintenance of pain thresholds and antianxiety behaviors under normal conditions, while silencing or suppressing the dmPFC/vlPAG pathway might be involved in the initial stages and maintenance of chronic pain and the emergence of anxiety-like behaviors.
Collapse
Affiliation(s)
- Jun-Bin Yin
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Department of Neurology, the 960th Hospital of PLA, Jinan, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA.,Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Shao-Hua Liang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Department of Human Anatomy, Binzhou Medical College, Yantai, China
| | - Fei Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Cadet Brigade, and
| | - Wen-Jun Zhao
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Cadet Brigade, and
| | - Yang Bai
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yi Sun
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Human Anatomy, Binzhou Medical College, Yantai, China
| | - Zhen-Yu Wu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | | | - Hai-Xia Liu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Ya-Cheng Lu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Jing Huang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhou-Feng Chen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jing Cao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Rui Ren
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Ya-Nan Peng
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Juan Yang
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Wei-Dong Zang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Xiang Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China.,Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
55
|
Lin R, Liang J, Luo M. The Raphe Dopamine System: Roles in Salience Encoding, Memory Expression, and Addiction. Trends Neurosci 2021; 44:366-377. [PMID: 33568331 DOI: 10.1016/j.tins.2021.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Dopamine (DA) neurons of the dorsal raphe nucleus (DRN) were traditionally viewed as an extension of the ventral tegmental area (VTA) DA population. While the VTA DA population is known to play important roles in reward processing, emerging evidence now supports the view that DRN DA neurons are a specialized midbrain DA subsystem that performs distinct functions in parallel to the VTA DA population. Recent studies have shed new light on the roles of DRN DA neurons in encoding incentive salience and in regulating memory expression and arousal. Here, we review recent findings using mouse models about the physiology and behavioral functions of DRN DA neurons, highlight the engagement of DRN DA neurons and their upstream circuits in opioid addiction, and discuss emerging lines of investigation that reveal multifaceted heterogeneity among DRN DA neurons.
Collapse
Affiliation(s)
- Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing 102206, China.
| | - Jingwen Liang
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Chinese Institute for Brain Research, Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
56
|
Sheng HY, Lv SS, Cai YQ, Shi W, Lin W, Liu TT, Lv N, Cao H, Zhang L, Zhang YQ. Activation of ventrolateral orbital cortex improves mouse neuropathic pain-induced anxiodepression. JCI Insight 2020; 5:e133625. [PMID: 33004687 PMCID: PMC7566721 DOI: 10.1172/jci.insight.133625] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Depression and anxiety are frequently observed in patients suffering from neuropathic pain. The underlying mechanisms remained unclear. The ventrolateral orbital cortex (VLO) has attracted considerable interest in its role in antidepressive effect in rodents. In the present study, we further investigated the role of the VLO in the anxiodepressive consequences of neuropathic pain in a chronic constriction injury of infraorbital nerve-induced trigeminal neuralgia (TN) mouse model. Elevated plus maze, open field, forced swimming, tail suspension, and sucrose preference tests were used to evaluate anxiodepressive-like behaviors. The results show that chemogenetic activation of bilateral VLO neurons, especially CaMK2A+ pyramidal neurons, blocked the TN-induced anxiodepressive-like behaviors. Chemogenetic and optogenetic activation of VGLUT2+ or inhibition of VGAT+ VLO neurons was sufficient to produce an antianxiodepressive effect in TN mice. Pharmacological activation of D1-like receptors (D1Rs) but not D2Rs in the VLO significantly alleviated TN-induced depressive-like behaviors. Electrophysiological recordings revealed a decreased excitability of VLO excitatory neurons following neuropathic pain. Furthermore, activation of submedius thalamic nucleus-VLO (Sm-VLO) projection mimicked the antianxiodepressive effect of VLO excitation. Conversely, activation of VLO-periaqueductal gray matter (PAG) projection had no effect on TN-induced anxiodepressive behaviors. This study provides a potentially novel mechanism-based therapeutic strategy for the anxiodepressive consequences of neuropathic pain.
Collapse
Affiliation(s)
- Hai-Yan Sheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
- Department of Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Henan, China
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ya-Qi Cai
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wu Shi
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, Shanghai, China
| | - Wei Lin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ting-Ting Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ning Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ling Zhang
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
57
|
Waung MW, Margolis EB, Charbit AR, Fields HL. A Midbrain Circuit that Mediates Headache Aversiveness in Rats. Cell Rep 2020; 28:2739-2747.e4. [PMID: 31509737 PMCID: PMC6831085 DOI: 10.1016/j.celrep.2019.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 06/07/2019] [Accepted: 07/24/2019] [Indexed: 01/05/2023] Open
Abstract
Migraines are a major health burden, but treatment is limited because of inadequate understanding of neural mechanisms underlying headache. Imaging studies of migraine patients demonstrate changes in both pain-modulatory circuits and reward-processing regions, but whether these changes contribute to the experience of headache is unknown. Here, we demonstrate a direct connection between the ventrolateral periaqueductal gray (vlPAG) and the ventral tegmental area (VTA) that contributes to headache aversiveness in rats. Many VTA neurons receive monosynaptic input from the vlPAG, and cranial nociceptive input increases Fos expression in VTA-projecting vlPAG neurons. Activation of PAG inputs to the VTA induces avoidance behavior, while inactivation of these projections induces a place preference only in animals with headache. This work identifies a distinct pathway that mediates cranial nociceptive aversiveness. Migraine headache is a common and debilitating disorder, yet its brain-activation patterns are poorly understood. Waung et al. discover that headache activates a connection between the periaqueductal gray and the ventral tegmental area in rats. Turning off this connection has no effect normally but decreases unpleasantness during headaches.
Collapse
Affiliation(s)
- Maggie W Waung
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Elyssa B Margolis
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Annabelle R Charbit
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Howard L Fields
- Alcohol and Addiction Research Group, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
58
|
Marciante AB, Farmer GE, Cunningham JT. G q DREADD activation of CaMKIIa MnPO neurons stimulates nitric oxide activity. J Neurophysiol 2020; 124:591-609. [PMID: 32697679 PMCID: PMC7500373 DOI: 10.1152/jn.00239.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 11/22/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) modify cellular activity following administration of the exogenous ligand clozapine-N-oxide (CNO). However, some reports indicate CNO may have off-target effects. The current studies investigate the use of Gq DREADDs in CaMKIIa-expressing neurons in the median preoptic nucleus (MnPO). Male Sprague-Dawley rats (250 g) anesthetized with isoflurane were stereotaxically microinjected in the MnPO with the Gq DREADD (AAV5-CaMKIIa-HM3D-mCherry) or control virus (AAV5-CaMKIIa-mCherry). Following a 2-wk recovery, rats were used for either immunohistochemical Fos analysis or in vitro patch-clamp electrophysiology. In Gq DREADD-injected rats, CNO induced significant increases in Fos staining in the MnPO and in regions that receive direct or indirect projections from the MnPO. In electrophysiological studies, CNO depolarized and augmented firing frequency in both Gq DREADD-positive neurons (Gq DREADD) as well as unlabeled MnPO neurons in slices from Gq DREADD-injected rats (Gq DREADDx). Gq DREADDx neurons also displayed increases in spontaneous postsynaptic current (sPSC) frequency in response to CNO. Additionally, CaMKIIa-positive MnPO neurons, which also express nitric oxide synthase (NOS), were treated with Nω-nitro-l-arginine (l-NNA; competitive inhibitor of NOS) and hemoglobin (NO scavenger) to assess the role of NO in Gq DREADDx neuron recruitment. Both l-NNA and hemoglobin blocked CNO-induced effects in Gq DREADDx neurons without affecting Gq DREADD neurons. These findings indicate that Gq DREADD-mediated activation of CaMKIIa/NOS expressing neurons in the MnPO can influence the activity of neighboring neurons. Future studies utilizing the use of Gq DREADDs will need to consider the potential recruitment of additional cell populations.NEW & NOTEWORTHY Rats were injected in the median preoptic nucleus (MnPO) with either an adeno-associated virus (AAV) and excitatory (Gq) designer receptor exclusively activated by designer drugs (DREADD) construct or a control AAV. In the Gq DREADD-injected rats only, clozapine-N-oxide (CNO) increased Fos staining in the MnPO and its targets and increased neuron action potential frequency. In electrophysiology experiments with slices with DREADD cells, unlabeled cells were activated and this was likely due to nitric oxide release by the DREADD cells.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Fort Worth, Texas
| | | |
Collapse
|
59
|
Ozcan S, Bulmus O, Ulker N, Canpolat S, Etem EO, Oruc S, Yardimci A, Bulmus FG, Ayar A, Kelestimur H, Ozcan M. Agomelatine potentiates anti-nociceptive effects of morphine in a mice model for diabetic neuropathy: involvement of NMDA receptor subtype NR1 within the raphe nucleus and periaqueductal grey. Neurol Res 2020; 42:554-563. [PMID: 32336224 DOI: 10.1080/01616412.2020.1757895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Opioid analgesics have been used for a long time in the treatment of acute and chronic pain. However, they have many side effects including tolerance development to a significant extent. Agomelatine, an atypical antidepressant, has been demonstrated to be effective in experimental studies on pain. However, the effect of agomelatine on morphine tolerance development and its mechanism of action are unknown. The antinociceptive effects of agomelatine, morphine and their combination were assessed in a mice model for painful diabetic neuropathy. The roles of glutamate ionotropic receptor N-methyl-D-aspartate (NMDA) type subunit-1 (GluN1) in raphe nucleus and periaqueductal gray (PAG) in the effect of agomelatine on neuropathic pain were also investigated in diabetic mice. METHODS Agomelatine (10 mg/kg), morphine (10 mg/kg) and agomelatine + morphine were administered intraperitoneally for 15 consecutive days (twice per day), and the analgesic responses were assessed at days 1, 3, 6, 9, 12 and 15 in healthy and diabetic mice. Real time polymerase chain reaction (RT-PCR) was used to determine the changes in GluN1 expression. RESULTS The tolerance development for morphine was evident, started at 6th day and remained thereafter, but not for agomelatine. GluN1 in raphe nucleus and PAG was upregulated in morphine treated but not in agomelatine-treated groups. DISCUSSION The combination of agomelatine with morphine alone causes outlasting analgesic effects of repeated treatment, which can be interpreted as attenuated tolerance. Moreover, we also pointed out for the first time the modulatory effects of agomelatine on GluN1 expression in raphe nucleus and PAG after chronic morphine treatment. ABBREVIATIONS Ca2+: Calcium; D2DR: Dopamine D2 receptor; GAPDH: Glyceraldehyde-3-phosphate dehydrogenase; GluN1: Glutamate ionotropic receptor N-methyl-D-aspartate type subunit-1; 5-HT: 5-hydroxytryptamine; i.p.: intraperitoneal injection; MPE: Maximal possible effect; MT: Melatonin; NMDA: N-methyl-D-aspartate; NMDAR1: NMDA receptors-1; PAG: Periaqueductal grey; PKCγ: Protein kinase C gamma; RT-PCR: Real time polymerase chain reaction; STZ: Streptozotocin.
Collapse
Affiliation(s)
- Sibel Ozcan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Ozgur Bulmus
- Department of Physiology, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Nazife Ulker
- Department of Physiology, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Sinan Canpolat
- Department of Physiology, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Ebru Onalan Etem
- Department of Medical Biology, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Sinem Oruc
- Department of Biophysics, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Ahmet Yardimci
- Department of Physiology, Faculty of Medicine, Firat University , Elazig, Turkey
| | | | - Ahmet Ayar
- Department of Physiology, Faculty of Medicine, Karadeniz Technical University , Trabzon, Turkey
| | - Haluk Kelestimur
- Department of Physiology, Faculty of Medicine, Firat University , Elazig, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University , Elazig, Turkey
| |
Collapse
|
60
|
Delery EC, Edwards S. Neuropeptide and cytokine regulation of pain in the context of substance use disorders. Neuropharmacology 2020; 174:108153. [PMID: 32470337 DOI: 10.1016/j.neuropharm.2020.108153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Substance use disorders (SUDs) are frequently accompanied by affective symptoms that promote negative reinforcement mechanisms contributing to SUD maintenance or progression. Despite their widespread use as analgesics, chronic or excessive exposure to alcohol, opioids, and nicotine produces heightened nociceptive sensitivity, termed hyperalgesia. This review focuses on the contributions of neuropeptide (CRF, melanocortin, opioid peptide) and cytokine (IL-1β, TNF-α, chemokine) systems in the development and maintenance of substance-induced hyperalgesia. Few effective therapies exist for either chronic pain or SUD, and the common interaction of these disease states likely complicates their effective treatment. Here we highlight promising new discoveries as well as identify gaps in research that could lead to more effective and even simultaneous treatment of SUDs and co-morbid hyperalgesia symptoms.
Collapse
Affiliation(s)
- Elizabeth C Delery
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
61
|
Neuroprotective and anti-inflammatory activity of DAT inhibitor R-phenylpiracetam in experimental models of inflammation in male mice. Inflammopharmacology 2020; 28:1283-1292. [PMID: 32279140 DOI: 10.1007/s10787-020-00705-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
R-phenylpiracetam (R-PhP, (4R)-2-(4-phenyl-2-oxopyrrolidin-1-yl)acetamide) is an optical isomer of phenotropil, a clinically-used nootropic drug that improves physical condition and cognition. Recently, R-PhP was shown to bind to the dopamine transporter (DAT). Since growing evidence suggests that dysfunction of the dopaminergic system is associated with persistent neuroinflammation, the aim of this study was to determine whether R-PhP, an inhibitor of DAT, has neuroprotective and anti-inflammatory effects in male mice. The pharmacokinetic profiles of R-PhP in mouse plasma and its bioavailability in brain tissue were assessed. To study possible molecular mechanisms involved in the anti-inflammatory activity of R-PhP, target profiling was performed using radioligand binding and enzymatic activity assays. To clarify the neuroprotective and anti-inflammatory effects of R-PhP, we used a lipopolysaccharide (LPS)-induced endotoxaemia model characterized by reduced body temperature and overexpression of inflammatory genes in the brain. In addition, the antinociceptive and anti-inflammatory effects of R-PhP were tested using carrageenan-induced paw oedema and formalin-induced paw-licking tests. R-PhP (50 mg/kg) reached the brain tissue 15 min after intraperitoneal (ip) and peroral (po) injections. The maximal concentration of R-PhP in the brain tissues was 28 µg/g and 18 µg/g tissue after ip and po administration, respectively. In radioligand binding assays, DAT was the only significant molecular target found for R-PhP. A single ip injection of R-PhP significantly attenuated the LPS-induced body temperature reduction and the overexpression of inflammatory genes, such as tumour necrosis factor-α (TNF-α), interleukin 1 beta (IL-1β) and inducible nitric oxide synthase (iNOS). Seven-day po pretreatment with R-PhP dose-dependently reduced paw oedema and the antinociceptive response, as shown by the carrageenan-induced paw oedema test. In addition, R-PhP decreased the nociceptive response during the inflammatory phase in the formalin-induced paw-licking test. Our study showed that R-PhP possesses neuroprotective and anti-inflammatory effects, demonstrating the potential of DAT inhibitors as effective therapeutics.
Collapse
|
62
|
Role of 5-HT1A Receptor in the Anxiolytic-Relaxant Effects of Bergamot Essential Oil in Rodent. Int J Mol Sci 2020; 21:ijms21072597. [PMID: 32283606 PMCID: PMC7177770 DOI: 10.3390/ijms21072597] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022] Open
Abstract
The essential oil obtained by the fresh fruit of Citrus bergamia Risso et Poiteau is used worldwide in aromatherapy to reduce pain, facilitate sleep induction, and/or minimize the effects of stress-induced anxiety. Preclinical pharmacological data demonstrate that bergamot essential oil (BEO) modulates specific neurotransmissions and shows an anxiolytic-relaxant effect not superimposable to that of the benzodiazepine diazepam, suggesting that neurotransmissions, other than GABAergic, could be involved. Several studies on essential oils indicate a role for serotonergic (5-HT) neurotransmission in anxiety. Interestingly, among serotonergic receptors, the 5-HT1A subtype seems to play a key role in the control of anxiety. Here, we report that modulation of the 5-HT1A receptor by selective agonist ((±)8-OH-DPAT) or antagonist (WAY-100635) may influence some of the anxiolytic-relaxant effects of BEO in Open Field and Elevated Plus Maze tests.
Collapse
|
63
|
Vaaga CE, Brown ST, Raman IM. Cerebellar modulation of synaptic input to freezing-related neurons in the periaqueductal gray. eLife 2020; 9:e54302. [PMID: 32207681 PMCID: PMC7124251 DOI: 10.7554/elife.54302] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/24/2020] [Indexed: 01/23/2023] Open
Abstract
Innate defensive behaviors, such as freezing, are adaptive for avoiding predation. Freezing-related midbrain regions project to the cerebellum, which is known to regulate rapid sensorimotor integration, raising the question of cerebellar contributions to freezing. Here, we find that neurons of the mouse medial (fastigial) cerebellar nuclei (mCbN), which fire spontaneously with wide dynamic ranges, send glutamatergic projections to the ventrolateral periaqueductal gray (vlPAG), which contains diverse cell types. In freely moving mice, optogenetically stimulating glutamatergic vlPAG neurons that express Chx10 reliably induces freezing. In vlPAG slices, mCbN terminals excite ~20% of neurons positive for Chx10 or GAD2 and ~70% of dopaminergic TH-positive neurons. Stimulating either mCbN afferents or TH neurons augments IPSCs and suppresses EPSCs in Chx10 neurons by activating postsynaptic D2 receptors. The results suggest that mCbN activity regulates dopaminergic modulation of the vlPAG, favoring inhibition of Chx10 neurons. Suppression of cerebellar output may therefore facilitate freezing.
Collapse
Affiliation(s)
| | - Spencer T Brown
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Indira M Raman
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
64
|
St Laurent R, Martinez Damonte V, Tsuda AC, Kauer JA. Periaqueductal Gray and Rostromedial Tegmental Inhibitory Afferents to VTA Have Distinct Synaptic Plasticity and Opiate Sensitivity. Neuron 2020; 106:624-636.e4. [PMID: 32191871 DOI: 10.1016/j.neuron.2020.02.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/01/2019] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
Abstract
The ventral tegmental area (VTA) is a major target of addictive drugs and receives multiple GABAergic projections originating outside the VTA. We describe differences in synaptic plasticity and behavior when optogenetically driving two opiate-sensitive GABAergic inputs to the VTA, the rostromedial tegmental nucleus (RMTg), and the periaqueductal gray (PAG). Activation of GABAergic RMTg terminals in the VTA in vivo is aversive, and low-frequency stimulation induces long-term depression in vitro. Low-frequency stimulation of PAG afferents in vitro unexpectedly causes long-term potentiation. Opioid receptor activation profoundly depresses PAG and RMTg inhibitory synapses but prevents synaptic plasticity only at PAG synapses. Activation of the GABAergic PAG terminals in the VTA promotes immobility, and optogenetically-driven immobility is blocked by morphine. Our data reveal the PAG as a source of highly opioid-sensitive GABAergic afferents and support the idea that different GABAergic pathways to the VTA control distinct behaviors.
Collapse
Affiliation(s)
- Robyn St Laurent
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Ayumi C Tsuda
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA.
| |
Collapse
|
65
|
van Reij RR, Joosten EA, van den Hoogen NJ. Dopaminergic neurotransmission and genetic variation in chronification of post-surgical pain. Br J Anaesth 2019; 123:853-864. [DOI: 10.1016/j.bja.2019.07.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/10/2019] [Accepted: 07/26/2019] [Indexed: 01/30/2023] Open
|
66
|
Cardozo Pinto DF, Yang H, Pollak Dorocic I, de Jong JW, Han VJ, Peck JR, Zhu Y, Liu C, Beier KT, Smidt MP, Lammel S. Characterization of transgenic mouse models targeting neuromodulatory systems reveals organizational principles of the dorsal raphe. Nat Commun 2019; 10:4633. [PMID: 31604921 PMCID: PMC6789139 DOI: 10.1038/s41467-019-12392-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/09/2019] [Indexed: 11/17/2022] Open
Abstract
The dorsal raphe (DR) is a heterogeneous nucleus containing dopamine (DA), serotonin (5HT), γ-aminobutyric acid (GABA) and glutamate neurons. Consequently, investigations of DR circuitry require Cre-driver lines that restrict transgene expression to precisely defined cell populations. Here, we present a systematic evaluation of mouse lines targeting neuromodulatory cells in the DR. We find substantial differences in specificity between lines targeting DA neurons, and in penetrance between lines targeting 5HT neurons. Using these tools to map DR circuits, we show that populations of neurochemically distinct DR neurons are arranged in a stereotyped topographical pattern, send divergent projections to amygdala subnuclei, and differ in their presynaptic inputs. Importantly, targeting DR DA neurons using different mouse lines yielded both structural and functional differences in the neural circuits accessed. These results provide a refined model of DR organization and support a comparative, case-by-case evaluation of the suitability of transgenic tools for any experimental application.
Collapse
Affiliation(s)
- Daniel F Cardozo Pinto
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongbin Yang
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Iskra Pollak Dorocic
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Johannes W de Jong
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Vivian J Han
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - James R Peck
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Yichen Zhu
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Christine Liu
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Kevin T Beier
- Departments of Physiology and Biophysics, Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, 92697, USA
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, FNWI University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Lammel
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|