1
|
Yang S, Wang D, Wang H, Shao J, Tang C. Design, synthesis, and biological evaluation of TRPV4-KCa2.3 coupling enhancers as novel therapeutic agents for hypertension. Eur J Med Chem 2025; 294:117724. [PMID: 40409057 DOI: 10.1016/j.ejmech.2025.117724] [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: 03/24/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/25/2025]
Abstract
Although current treatment strategies for hypertension are well-developed, there remains a group of patients who do not respond adequately to available medications. As a result, the identification of new therapeutic targets and the design of target-specific drugs are crucial directions for the future management of hypertension. Our previous research identified the TRPV4-KCa2.3 complex as a novel target for hypertension treatment, leading to the discovery of the positive compound JNc-440. Using JNc-440 as a lead molecule, 21 compounds were designed and synthesized across five distinct series. Among these, representative compounds IB-2 and II-9 demonstrated the ability to restore the coupling of the decoupled complex under hypertensive conditions and significantly reduced blood pressure in a high salt-induced hypertensive mouse model. This work lays a foundation for the future development of novel therapeutics targeting hypertension.
Collapse
Affiliation(s)
- Shaying Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongyu Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Huabing Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Junlan Shao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China.
| |
Collapse
|
2
|
Markovic T, Higginbotham J, Ruyle B, Massaly N, Yoon HJ, Kuo CC, Kim JR, Yi J, Garcia JJ, Sze E, Abt J, Teich RH, Dearman JJ, McCall JG, Morón JA. A locus coeruleus to dorsal hippocampus pathway mediates cue-induced reinstatement of opioid self-administration in male and female rats. Neuropsychopharmacology 2024; 49:915-923. [PMID: 38374364 PMCID: PMC11039689 DOI: 10.1038/s41386-024-01828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
Opioid use disorder is a chronic relapsing disorder encompassing misuse, dependence, and addiction to opioid drugs. Long term maintenance of associations between the reinforcing effects of the drug and the cues associated with its intake are a leading cause of relapse. Indeed, exposure to the salient drug-associated cues can lead to drug cravings and drug seeking behavior. The dorsal hippocampus (dHPC) and locus coeruleus (LC) have emerged as important structures for linking the subjective rewarding effects of opioids with environmental cues. However, their role in cue-induced reinstatement of opioid use remains to be further elucidated. In this study, we showed that chemogenetic inhibition of excitatory dHPC neurons during re-exposure to drug-associated cues significantly attenuates cue-induced reinstatement of morphine-seeking behavior. In addition, the same manipulation reduced reinstatement of sucrose-seeking behavior but failed to alter memory recall in the object location task. Finally, intact activity of tyrosine hydroxylase (TH) LC-dHPCTh afferents is necessary to drive cue induced reinstatement of morphine-seeking as inhibition of this pathway blunts cue-induced drug-seeking behavior. Altogether, these studies show an important role of the dHPC and LC-dHPCTh pathway in mediating cue-induced reinstatement of opioid seeking.
Collapse
Affiliation(s)
- Tamara Markovic
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica Higginbotham
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Brian Ruyle
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Hye Jean Yoon
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jenny R Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jiwon Yi
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jeniffer J Garcia
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric Sze
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Julian Abt
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Rachel H Teich
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Joanna J Dearman
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
- Pain Center, Washington University in St Louis, St. Louis, MO, USA
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis, St. Louis, MO, USA
| | - Jose A Morón
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA.
- Pain Center, Washington University in St Louis, St. Louis, MO, USA.
- School of Medicine, Washington University in St Louis, St. Louis, MO, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
3
|
Zhang Y, Shaabani S, Vowinkel K, Trombetta-Lima M, Sabogal-Guáqueta AM, Chen T, Hoekstra J, Lembeck J, Schmidt M, Decher N, Dömling A, Dolga AM. Novel SK channel positive modulators prevent ferroptosis and excitotoxicity in neuronal cells. Biomed Pharmacother 2024; 171:116163. [PMID: 38242037 DOI: 10.1016/j.biopha.2024.116163] [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: 10/23/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Small conductance calcium-activated potassium (SK) channel activity has been proposed to play a role in the pathology of several neurological diseases. Besides regulating plasma membrane excitability, SK channel activation provides neuroprotection against ferroptotic cell death by reducing mitochondrial Ca2+ uptake and reactive oxygen species (ROS). In this study, we employed a multifaceted approach, integrating structure-based and computational techniques, to strategically design and synthesize an innovative class of potent small-molecule SK2 channel modifiers through highly efficient multicomponent reactions (MCRs). The compounds' neuroprotective activity was compared with the well-studied SK positive modulator, CyPPA. Pharmacological SK channel activation by selected compounds confers neuroprotection against ferroptosis at low nanomolar ranges compared to CyPPA, that mediates protection at micromolar concentrations, as shown by an MTT assay, real-time cell impedance measurements and propidium iodide staining (PI). These novel compounds suppress increased mitochondrial ROS and Ca2+ level induced by ferroptosis inducer RSL3. Moreover, axonal degeneration was rescued by these novel SK channel activators in primary mouse neurons and they attenuated glutamate-induced neuronal excitability, as shown via microelectrode array. Meanwhile, functional afterhyperpolarization of the novel SK2 channel modulators was validated by electrophysiological measurements showing more current change induced by the novel modulators than the reference compound, CyPPA. These data support the notion that SK2 channel activation can represent a therapeutic target for brain diseases in which ferroptosis and excitotoxicity contribute to the pathology.
Collapse
Affiliation(s)
- Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Shabnam Shaabani
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technologies and Biopharmacy, Research Institute of Pharmacy, University of Groningen, the Netherlands
| | | | - Tingting Chen
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Hoekstra
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Jan Lembeck
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, 35037 Marburg, Germany
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, the Netherlands.
| |
Collapse
|
4
|
Ibrahim KM, Massaly N, Yoon HJ, Sandoval R, Widman AJ, Heuermann RJ, Williams S, Post W, Pathiranage S, Lintz T, Zec A, Park A, Yu W, Kash TL, Gereau RW, Morón JA. Dorsal hippocampus to nucleus accumbens projections drive reinforcement via activation of accumbal dynorphin neurons. Nat Commun 2024; 15:750. [PMID: 38286800 PMCID: PMC10825206 DOI: 10.1038/s41467-024-44836-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/04/2024] [Indexed: 01/31/2024] Open
Abstract
The hippocampus is pivotal in integrating emotional processing, learning, memory, and reward-related behaviors. The dorsal hippocampus (dHPC) is particularly crucial for episodic, spatial, and associative memory, and has been shown to be necessary for context- and cue-associated reward behaviors. The nucleus accumbens (NAc), a central structure in the mesolimbic reward pathway, integrates the salience of aversive and rewarding stimuli. Despite extensive research on dHPC→NAc direct projections, their sufficiency in driving reinforcement and reward-related behavior remains to be determined. Our study establishes that activating excitatory neurons in the dHPC is sufficient to induce reinforcing behaviors through its direct projections to the dorso-medial subregion of the NAc shell (dmNAcSh). Notably, dynorphin-containing neurons specifically contribute to dHPC-driven reinforcing behavior, even though both dmNAcSh dynorphin- and enkephalin-containing neurons are activated with dHPC stimulation. Our findings unveil a pathway governing reinforcement, advancing our understanding of the hippocampal circuity's role in reward-seeking behaviors.
Collapse
Affiliation(s)
- Khairunisa Mohamad Ibrahim
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
- Department of Anesthesiology and Perioperative Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Hye-Jean Yoon
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Rossana Sandoval
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Allie J Widman
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Robert J Heuermann
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University Pain Center, St. Louis, MO, 63110, USA
| | - Sidney Williams
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - William Post
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Sulan Pathiranage
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Tania Lintz
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Azra Zec
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Ashley Park
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
| | - Waylin Yu
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Robert W Gereau
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Jose A Morón
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO, 63110, USA.
- Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
5
|
Wu X, Yan Q, Liu L, Xue X, Yao W, Li X, Li W, Ding S, Xia Y, Zhang D, Zhu F. Domesticated HERV-W env contributes to the activation of the small conductance Ca 2+-activated K + type 2 channels via decreased 5-HT4 receptor in recent-onset schizophrenia. Virol Sin 2023; 38:9-22. [PMID: 36007838 PMCID: PMC10006216 DOI: 10.1016/j.virs.2022.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
The human endogenous retroviruses type W family envelope (HERV-W env) gene is located on chromosome 7q21-22. Our previous studies show that HERV-W env is elevated in schizophrenia and HERV-W env can increase calcium influx. Additionally, the 5-HTergic system and particularly 5-hydroxytryptamine (5-HT) receptors play a prominent role in the pathogenesis and treatment of schizophrenia. 5-hydroxytryptamine receptor 4 (5-HT4R) agonist can block calcium channels. However, the underlying relationship between HERV-W env and 5-HT4R in the etiology of schizophrenia has not been revealed. Here, we used enzyme-linked immunosorbent assay to detect the concentration of HERV-W env and 5-HT4R in the plasma of patients with schizophrenia and we found that there were decreased levels of 5-HT4R and a negative correlation between 5-HT4R and HERV-W env in schizophrenia. Overexpression of HERV-W env decreased the transcription and protein levels of 5-HT4R but increased small conductance Ca2+-activated K+ type 2 channels (SK2) expression levels. Further studies revealed that HERV-W env could interact with 5-HT4R. Additionally, luciferase assay showed that an essential region (-364 to -176 from the transcription start site) in the SK2 promoter was required for HERV-W env-induced SK2 expression. Importantly, 5-HT4R participated in the regulation of SK2 expression and promoter activity. Electrophysiological recordings suggested that HERV-W env could increase SK2 channel currents and the increase of SK2 currents was inhibited by 5-HT4R. In conclusion, HERV-W env could activate SK2 channels via decreased 5-HT4R, which might exhibit a novel mechanism for HERV-W env to influence neuronal activity in schizophrenia.
Collapse
Affiliation(s)
- Xiulin Wu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Qiujin Yan
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | | | - Xing Xue
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei Yao
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xuhang Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenshi Li
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuang Ding
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yaru Xia
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Dongyan Zhang
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Fan Zhu
- State Key Laboratory of Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China; Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
6
|
Zahra A, Liu R, Han W, Meng H, Wang Q, Wang Y, Campbell SL, Wu J. K Ca-Related Neurological Disorders: Phenotypic Spectrum and Therapeutic Indications. Curr Neuropharmacol 2023; 21:1504-1518. [PMID: 36503451 PMCID: PMC10472807 DOI: 10.2174/1570159x21666221208091805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Although potassium channelopathies have been linked to a wide range of neurological conditions, the underlying pathogenic mechanism is not always clear, and a systematic summary of clinical manifestation is absent. Several neurological disorders have been associated with alterations of calcium-activated potassium channels (KCa channels), such as loss- or gain-of-function mutations, post-transcriptional modification, etc. Here, we outlined the current understanding of the molecular and cellular properties of three subtypes of KCa channels, including big conductance KCa channels (BK), small conductance KCa channels (SK), and the intermediate conductance KCa channels (IK). Next, we comprehensively reviewed the loss- or gain-of-function mutations of each KCa channel and described the corresponding mutation sites in specific diseases to broaden the phenotypic-genotypic spectrum of KCa-related neurological disorders. Moreover, we reviewed the current pharmaceutical strategies targeting KCa channels in KCa-related neurological disorders to provide new directions for drug discovery in anti-seizure medication.
Collapse
Affiliation(s)
- Aqeela Zahra
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan
| | - Ru Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Wenzhe Han
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Hui Meng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Qun Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - YunFu Wang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Susan L. Campbell
- Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
7
|
Luján R, Merchán-Pérez A, Soriano J, Martín-Belmonte A, Aguado C, Alfaro-Ruiz R, Moreno-Martínez AE, DeFelipe J. Neuron Class and Target Variability in the Three-Dimensional Localization of SK2 Channels in Hippocampal Neurons as Detected by Immunogold FIB-SEM. Front Neuroanat 2022; 15:781314. [PMID: 34975419 PMCID: PMC8715088 DOI: 10.3389/fnana.2021.781314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022] Open
Abstract
Small-conductance calcium-activated potassium (SK) channels are crucial for learning and memory. However, many aspects of their spatial organization in neurons are still unknown. In this study, we have taken a novel approach to answering these questions combining a pre-embedding immunogold labeling with an automated dual-beam electron microscope that integrates focused ion beam milling and scanning electron microscopy (FIB/SEM) to gather 3D map ultrastructural and biomolecular information simultaneously. Using this new approach, we evaluated the number and variability in the density of extrasynaptic SK2 channels in 3D reconstructions from six dendritic segments of excitatory neurons and six inhibitory neurons present in the stratum radiatum of the CA1 region of the mouse. SK2 immunoparticles were observed throughout the surface of hippocampal neurons, either scattered or clustered, as well as at intracellular sites. Quantitative volumetric evaluations revealed that the extrasynaptic SK2 channel density in spines was seven times higher than in dendritic shafts and thirty-five times higher than in interneurons. Spines showed a heterogeneous population of SK2 expression, some spines having a high SK2 content, others having a low content and others lacking SK2 channels. SK2 immunonegative spines were significantly smaller than those immunopositive. These results show that SK2 channel density differs between excitatory and inhibitory neurons and demonstrates a large variability in the density of SK2 channels in spines. Furthermore, we demonstrated that SK2 expression was associated with excitatory synapses, but not with inhibitory synapses in CA1 pyramidal cells. Consequently, regulation of excitability and synaptic plasticity by SK2 channels is expected to be neuron class- and target-specific. These data show that immunogold FIB/SEM represent a new powerful EM tool to correlate structure and function of ion channels with nanoscale resolution.
Collapse
Affiliation(s)
- Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Angel Merchán-Pérez
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Joaquim Soriano
- CRIB-Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Ana Esther Moreno-Martínez
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain.,Instituto Cajal (CSIC), Madrid, Spain
| |
Collapse
|
8
|
Shang Q, Xiao J, Gao B, Liang M, Wang J, Qian H, Xi Z, Li T, Liu X. D1R/PP2A/p-CaMKIIα signaling in the caudate putamen is involved in acute methamphetamine-induced hyperlocomotion. Neurosci Lett 2021; 760:136102. [PMID: 34237414 DOI: 10.1016/j.neulet.2021.136102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
Drug addiction is underscored by the transition from experimental use to dependent use of addictive drugs. Acute use of methamphetamine (METH) causes a range of clinical symptoms, including hyperlocomotion. Dopamine D1 receptor (D1R)-mediated negative regulation of phosphorylated calcium/calmodulin-dependent protein kinase IIα (p-CaMKIIα, threonine [Thr] 286) is involved in the acute effects induced by single METH administration. Protein phosphatase 2A (PP2A) is a potential bridge that links D1R and p-CaMKIIα (Thr 286) after acute METH administration. However, the mechanisms underlying hyperlocomotion induced by single METH administration remain unclear. In this study, SCH23390 (a D1R inhibitor) and LB100 (a PP2A inhibitor) were administered to examine the involvement of D1R and PP2A signaling in acute METH-induced hyperlocomotion in mice. The protein levels of methylated PP2A-C (m-PP2A-C, leucine [Leu] 309), phosphorylated PP2A-C (p-PP2A-C, tyrosine [Tyr] 307), PP2A-C, p-CaMKIIα (Thr 286), and CaMKIIα in the prefrontal cortex (PFc), nucleus accumbens (NAc), and caudate putamen (CPu) were measured. Administration of 0.5 mg/kg SCH23390 reversed the acute METH-induced increase in protein levels of m-PP2A-C (Leu 309) and the decrease in protein levels of p-PP2A-C (Tyr 307) in the CPu, but not in the PFC and NAc. Moreover, prior administration of 0.1 mg/kg LB100 attenuated hyperlocomotion induced by single METH administration and reversed the decrease in protein levels of p-CaMKII (Thr 286) in the PFC, NAc, and CPu. Collectively, these results indicate that the D1R/PP2A/p-CaMKIIα signaling cascade in the CPu may be involved in hyperlocomotion after a single administration of METH.
Collapse
Affiliation(s)
- Qing Shang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Jing Xiao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Baoyao Gao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Min Liang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Jing Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Hongyan Qian
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Zhijia Xi
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China
| | - Tao Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China.
| | - Xinshe Liu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, People's Republic of China; Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, People's Republic of China.
| |
Collapse
|
9
|
Shang Q, Liang M, Xiao J, Gao B, Qian H, Wang J, Chen G, Fang J, Li T, Liu X. LB100 attenuates methamphetamine-induced behavioral sensitization by inhibiting the Raf1-ERK 1/2 cascade in the caudate putamen. Neuroreport 2021; 32:988-993. [PMID: 34102646 DOI: 10.1097/wnr.0000000000001678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Methamphetamine (METH) abuse has become a serious social problem. Behavioral sensitization is a common behavioral paradigm used to study the neurobiological mechanism that underlies drug addiction. Our previous study demonstrated that the activity of protein phosphatase 2A (PP2A) and the level of phosphorylated extracellular signal-related kinase 1/2 (p-ERK 1/2) are increased in the caudate putamen (CPu) of METH-sensitive mice. However, the relationship between PP2A and ERK 1/2 in METH-induced behavioral sensitization remains unknown. Some studies have indicated that Raf1 may be involved in this process. In this study, LB100, a PP2A inhibitor for treating solid tumors, was first used to clarify the relationship between PP2A and ERK 1/2. In addition, Western blot was used to examine the levels of p-Raf1 (Ser 259) and p-ERK 1/2 (Thr 202/Tyr 204) in the CPu, hippocampus (Hip) and nucleus accumbens (NAc). Our results showed that 2 mg/kg LB100 significantly attenuated METH-induced behavioral sensitization. Furthermore, Western blot analysis revealed that pretreatment with 2 mg/kg LB100 remarkably reversed METH-induced reduction of p-Raf1, as well as upregulation of p-ERK 1/2 in the CPu. Taken together, these results indicate that PP2A plays an important role in METH-induced behavioral sensitization and phosphorylates ERK 1/2 by dephosphorylating p-Raf1 in the CPu to further regulate METH-induced behavioral sensitization.
Collapse
Affiliation(s)
- Qing Shang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Min Liang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jing Xiao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Baoyao Gao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Hongyan Qian
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jing Wang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Gang Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jie Fang
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Tao Li
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Xinshe Liu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center.,Institute of Forensic Injury, Institute of Forensic Bioevidence, Western China Science and Technology Innovation Harbor, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
10
|
McCoy MT, Jayanthi S, Cadet JL. Potassium Channels and Their Potential Roles in Substance Use Disorders. Int J Mol Sci 2021; 22:1249. [PMID: 33513859 PMCID: PMC7865894 DOI: 10.3390/ijms22031249] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 01/12/2023] Open
Abstract
Substance use disorders (SUDs) are ubiquitous throughout the world. However, much remains to be done to develop pharmacotherapies that are very efficacious because the focus has been mostly on using dopaminergic agents or opioid agonists. Herein we discuss the potential of using potassium channel activators in SUD treatment because evidence has accumulated to support a role of these channels in the effects of rewarding drugs. Potassium channels regulate neuronal action potential via effects on threshold, burst firing, and firing frequency. They are located in brain regions identified as important for the behavioral responses to rewarding drugs. In addition, their expression profiles are influenced by administration of rewarding substances. Genetic studies have also implicated variants in genes that encode potassium channels. Importantly, administration of potassium agonists have been shown to reduce alcohol intake and to augment the behavioral effects of opioid drugs. Potassium channel expression is also increased in animals with reduced intake of methamphetamine. Together, these results support the idea of further investing in studies that focus on elucidating the role of potassium channels as targets for therapeutic interventions against SUDs.
Collapse
Affiliation(s)
| | | | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, USA; (M.T.M.); (S.J.)
| |
Collapse
|
11
|
Sun J, Liu Y, Baudry M, Bi X. SK2 channel regulation of neuronal excitability, synaptic transmission, and brain rhythmic activity in health and diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118834. [PMID: 32860835 PMCID: PMC7541745 DOI: 10.1016/j.bbamcr.2020.118834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 11/20/2022]
Abstract
Small conductance calcium-activated potassium channels (SKs) are solely activated by intracellular Ca2+ and their activation leads to potassium efflux, thereby repolarizing/hyperpolarizing membrane potential. Thus, these channels play a critical role in synaptic transmission, and consequently in information transmission along the neuronal circuits expressing them. SKs are widely but not homogeneously distributed in the central nervous system (CNS). Activation of SKs requires submicromolar cytoplasmic Ca2+ concentrations, which are reached following either Ca2+ release from intracellular Ca2+ stores or influx through Ca2+ permeable membrane channels. Both Ca2+ sensitivity and synaptic levels of SKs are regulated by protein kinases and phosphatases, and degradation pathways. SKs in turn control the activity of multiple Ca2+ channels. They are therefore critically involved in coordinating diverse Ca2+ signaling pathways and controlling Ca2+ signal amplitude and duration. This review highlights recent advances in our understanding of the regulation of SK2 channels and of their roles in normal brain functions, including synaptic plasticity, learning and memory, and rhythmic activities. It will also discuss how alterations in their expression and regulation might contribute to various brain disorders such as Angelman Syndrome, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Michel Baudry
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
12
|
Shrestha A, Sultana R, Adeniyi PA, Lee CC, Ogundele OM. Positive Modulation of SK Channel Impedes Neuron-Specific Cytoskeletal Organization and Maturation. Dev Neurosci 2020; 42:59-71. [PMID: 32580196 PMCID: PMC7486235 DOI: 10.1159/000507989] [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: 07/31/2019] [Accepted: 04/15/2020] [Indexed: 01/01/2023] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) modulates the structural plasticity of dendritic spines by impacting cytoskeletal organization and kinase signaling. In the developing nervous system, activation of NMDAR is pertinent for neuronal migration, neurite differentiation, and cellular organization. Given that small conductance potassium channels (SK2/3) repress NMDAR ionotropic signaling, this study highlights the impact of neonatal SK channel potentiation on adult cortical and hippocampal organization. Neonatal SK channel potentiation was performed by one injection of SK2/3 agonist (CyPPA) into the pallium of mice on postnatal day 2 (P2). When the animals reached adulthood (P55), the hippocampus and cortex were examined to assess neuronal maturation, lamination, and the distribution of synaptic cytoskeletal proteins. Immunodetection of neuronal markers in the brain of P2-treated P55 mice revealed the presence of immature neurons in the upper cortical layers (layers II-IV) and CA1 (hippocampus). Also, layer-dependent cortical-cell density was attenuated due to the ectopic localization of mature (NeuN+) and immature (Doublecortin+ [DCX+]) neurons in cortical layers II-IV. Similarly, the decreased count of NeuN+ neurons in the CA1 is accompanied by an increase in the number of immature DCX+ neurons. Ectopic localization of neurons in the upper cortex and CA1 caused the dramatic expression of neuron-specific cytoskeletal proteins. In line with this, structural deformity of neuronal projections and the loss of postsynaptic densities suggests that postsynaptic integrity is compromised in the SK2/3+ brain. From these results, we deduced that SK channel activity in the developing brain likely impacts neuronal maturation through its effects on cytoskeletal formation.
Collapse
Affiliation(s)
- Amita Shrestha
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Philip A Adeniyi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Charles C Lee
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Olalekan M Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA,
| |
Collapse
|
13
|
Campos-Jurado Y, Martí-Prats L, Morón JA, Polache A, Granero L, Hipólito L. Dose-dependent induction of CPP or CPA by intra-pVTA ethanol: Role of mu opioid receptors and effects on NMDA receptors. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109875. [PMID: 31978422 PMCID: PMC7096259 DOI: 10.1016/j.pnpbp.2020.109875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 12/16/2022]
Abstract
The neurobiological mechanisms underlying alcohol motivational properties are still not fully understood, however, the mu-opioid receptors (MORs) have been evidenced as central elements in the manifestation of the alcohol reinforcing properties. Drug-associated environmental stimuli can trigger alcohol relapse and promote alcohol consumption whereby N-methyl-d-aspartate (NMDA) receptors play a pivotal role. Here we sought to demonstrate, for the first time, that ethanol induces conditioned place preference or aversion (CPP or CPA) when administered locally into the ventral tegmental area (VTA) and the associated role of MORs. We further analyzed the changes in the expression and mRNA levels of GluN1 and GluN2A subunits in designated brain areas. The expression of CPP or CPA was characterized following intra-VTA ethanol administration and we showed that either reinforcing (CPP) or aversive (CPA) properties are dependent on the dose administered (ranging here from 35 to 300 nmol). Furthermore, the critical contribution of local MORs in the acquisition of CPP was revealed by a selective antagonist, namely β-Funaltrexamine. Finally, modifications of the expression of NMDA receptor subunits in the Nucleus Accumbens (NAc) and Hippocampus after ethanol-induced CPP were analyzed at the proteomic and transcriptomic levels by western blot and In Situ Hybridation RNAscope techniques, respectively. Results showed that the mRNA levels of GluN2A but not GluN1 in NAc are higher after ethanol CPP. These novel results pave the way for further characterisation of the mechanisms by which ethanol motivational properties are associated with learned environmental cues.
Collapse
Affiliation(s)
- Yolanda Campos-Jurado
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Lucía Martí-Prats
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Jose A Morón
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ana Polache
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Luis Granero
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain
| | - Lucía Hipólito
- Department of Pharmacy and Pharmaceutical Tech. and Parasit., University of València, Spain.
| |
Collapse
|
14
|
Qu L, Wang Y, Li Y, Wang X, Li N, Ge S, Wang J, Wang GJ, Volkow ND, Lang B, Wang P, Wu H, Zeng J, Fu J, Li J, Zhang Y, Wang X. Decreased Neuronal Excitability in Medial Prefrontal Cortex during Morphine Withdrawal is associated with enhanced SK channel activity and upregulation of small GTPase Rac1. Am J Cancer Res 2020; 10:7369-7383. [PMID: 32641997 PMCID: PMC7330845 DOI: 10.7150/thno.44893] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Neuroadaptations in the medial prefrontal cortex (mPFC) and Nucleus Accumbens (NAc) play a role in the disruption of control-reward circuits in opioid addiction. Small Conductance Calcium-Activated Potassium (SK) channels in the mPFC have been implicated in neuronal excitability changes during morphine withdrawal. However, the mechanism that modulates SK channels during withdrawal is still unknown. Methods: Rats were exposed for one week to daily morphine injections (10 mg·kg-1 s.c.) followed by conditional place preference (CPP) assessment. One week after withdrawal, electrophysiological, morphological and molecular biological methods were applied to investigate the effects of morphine on SK channels in mPFC, including infralimbic (IL), prelimbic (PrL) cortices and NAc (core and shell). We verified the hypothesis that Rac1, a member of Rho family of small GTPases, implicated in SK channel regulation, modulate SK channel neuroadaptations during opiate withdrawal. Results: One week after morphine withdrawal, the neuronal excitability of layer 5 pyramidal neurons in IL was decreased, but not in PrL. Whereas, the excitability was increased in NAc-shell, but not in NAc-core. In mPFC, the expression of the SK3 subunit was enhanced after one-week of withdrawal compared to controls. In the IL, Rac1 signaling was increased during withdrawal, and the Rac1 inhibitor NSC23766 disrupted SK current, which increased neuronal firing. Suppression of Rac1 inhibited morphine-induced CPP and expression of SK channels in IL. Conclusions: These findings highlight the potential value of SK channels and the upstream molecule Rac1, which may throw light on the therapeutic mechanism of neuromodulation treatment for opioid dependence.
Collapse
|
15
|
CXCR7 regulates epileptic seizures by controlling the synaptic activity of hippocampal granule cells. Cell Death Dis 2019; 10:825. [PMID: 31672961 PMCID: PMC6823462 DOI: 10.1038/s41419-019-2052-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022]
Abstract
C–X–C motif chemokine receptor 7 (CXCR7), which mediates the immune response in the brain, was recently reported to regulate neurological functions. However, the role of CXCR7 in epilepsy remains unclear. Here, we found that CXCR7 was upregulated in the hippocampal dentate gyrus (DG) of mice subjected to kainic acid (KA)-induced epilepsy and in the brain tissues of patients with temporal lobe epilepsy. Silencing CXCR7 in the hippocampal DG region exerted an antiepileptic effect on the KA-induced mouse model of epilepsy, whereas CXCR7 overexpression produced a seizure-aggravating effect. Mechanistically, CXCR7 selectively regulated N-methyl-d-aspartate receptor (NMDAR)-mediated synaptic neurotransmission in hippocampal dentate granule cells by modulating the cell membrane expression of the NMDAR subunit2A, which requires the activation of extracellular signal-regulated kinase 1/2 (ERK1/2). Thus, CXCR7 may regulate epileptic seizures and represents a novel target for antiepileptic treatments.
Collapse
|
16
|
Fakira AK, Peck EG, Liu Y, Lueptow LM, Trimbake NA, Han MH, Calipari ES, Devi LA. The role of the neuropeptide PEN receptor, GPR83, in the reward pathway: Relationship to sex-differences. Neuropharmacology 2019; 157:107666. [PMID: 31199956 DOI: 10.1016/j.neuropharm.2019.107666] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/22/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
Abstract
GPR83, the receptor for the neuropeptide PEN, exhibits high expression in the nucleus accumbens of the human and rodent brain, suggesting that it plays a role in modulating the mesolimbic reward pathway. However, the cell-type specific expression of GPR83, its functional impact in the reward pathway, and in drug reward-learning has not been fully explored. Using GPR83/eGFP mice, we show high GPR83 expression on cholinergic interneurons in the nucleus accumbens and moderate expression on ventral tegmental area dopamine neurons. In GPR83 knockout mice, baseline dopamine release in the nucleus accumbens is enhanced which disrupts the ratio of tonic vs phasic release. Additionally, GPR83 knockout leads to changes in the expression of dopamine-related genes. Using the morphine conditioned place preference model, we identify sex differences in morphine reward-learning, show that GPR83 is upregulated in the nucleus accumbens following morphine conditioned place preference, and show that shRNA-mediated knockdown of GPR83 in the nucleus accumbens leads to attenuation morphine reward. Together, these findings detect GPR83 expression in the reward-pathway, and show its involvement in dopamine release and morphine reward-learning.
Collapse
Affiliation(s)
- Amanda K Fakira
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Emily G Peck
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yutong Liu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA; Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Lindsay M Lueptow
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Nikita A Trimbake
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA; Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, NY, NY, USA; Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY, NY, USA.
| |
Collapse
|
17
|
Grove J, Ripke S, Als TD, Mattheisen M, Walters RK, Won H, Pallesen J, Agerbo E, Andreassen OA, Anney R, Awashti S, Belliveau R, Bettella F, Buxbaum JD, Bybjerg-Grauholm J, Bækvad-Hansen M, Cerrato F, Chambert K, Christensen JH, Churchhouse C, Dellenvall K, Demontis D, De Rubeis S, Devlin B, Djurovic S, Dumont AL, Goldstein JI, Hansen CS, Hauberg ME, Hollegaard MV, Hope S, Howrigan DP, Huang H, Hultman CM, Klei L, Maller J, Martin J, Martin AR, Moran JL, Nyegaard M, Nærland T, Palmer DS, Palotie A, Pedersen CB, Pedersen MG, dPoterba T, Poulsen JB, Pourcain BS, Qvist P, Rehnström K, Reichenberg A, Reichert J, Robinson EB, Roeder K, Roussos P, Saemundsen E, Sandin S, Satterstrom FK, Davey Smith G, Stefansson H, Steinberg S, Stevens CR, Sullivan PF, Turley P, Walters GB, Xu X, Stefansson K, Geschwind DH, Nordentoft M, Hougaard DM, Werge T, Mors O, Mortensen PB, Neale BM, Daly MJ, Børglum AD. Identification of common genetic risk variants for autism spectrum disorder. Nat Genet 2019; 51:431-444. [PMID: 30804558 PMCID: PMC6454898 DOI: 10.1038/s41588-019-0344-8] [Citation(s) in RCA: 1431] [Impact Index Per Article: 238.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder (ASD) is a highly heritable and heterogeneous group of neurodevelopmental phenotypes diagnosed in more than 1% of children. Common genetic variants contribute substantially to ASD susceptibility, but to date no individual variants have been robustly associated with ASD. With a marked sample-size increase from a unique Danish population resource, we report a genome-wide association meta-analysis of 18,381 individuals with ASD and 27,969 controls that identified five genome-wide-significant loci. Leveraging GWAS results from three phenotypes with significantly overlapping genetic architectures (schizophrenia, major depression, and educational attainment), we identified seven additional loci shared with other traits at equally strict significance levels. Dissecting the polygenic architecture, we found both quantitative and qualitative polygenic heterogeneity across ASD subtypes. These results highlight biological insights, particularly relating to neuronal function and corticogenesis, and establish that GWAS performed at scale will be much more productive in the near term in ASD.
Collapse
Affiliation(s)
- Jakob Grove
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Stephan Ripke
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Thomas D Als
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Manuel Mattheisen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Raymond K Walters
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonatan Pallesen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Esben Agerbo
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Ole A Andreassen
- NORMENT-KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Richard Anney
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Swapnil Awashti
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin, Berlin, Germany
| | - Rich Belliveau
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Francesco Bettella
- NORMENT-KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonas Bybjerg-Grauholm
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kimberly Chambert
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jane H Christensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Claire Churchhouse
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Karin Dellenvall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Srdjan Djurovic
- NORMENT-KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ashley L Dumont
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jacqueline I Goldstein
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Christine S Hansen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Institute of Biological Psychiatry, MHC SctHans, Mental Health Services, Copenhagen, Denmark
| | - Mads Engel Hauberg
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Mads V Hollegaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Sigrun Hope
- NORMENT-KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
- Department of Neurohabilitation, Oslo University Hospital, Oslo, Norway
| | - Daniel P Howrigan
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Lambertus Klei
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julian Maller
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Genomics plc, Oxford, UK
- Vertex Pharmaceuticals, Abingdon, UK
| | - Joanna Martin
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jennifer L Moran
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mette Nyegaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | - Terje Nærland
- NORMENT-KG Jebsen Centre for Psychosis Research, University of Oslo, Oslo, Norway
- NevSom, Department of Rare Disorders and Disabilities, , Oslo University Hospital, Oslo, Norway
| | - Duncan S Palmer
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aarno Palotie
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Carsten Bøcker Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Marianne Giørtz Pedersen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Timothy dPoterba
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jesper Buchhave Poulsen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Beate St Pourcain
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Per Qvist
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark
| | | | - Abraham Reichenberg
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer Reichert
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elise B Robinson
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kathryn Roeder
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Statistics and Data Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
| | | | - Sven Sandin
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - F Kyle Satterstrom
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | | | - Christine R Stevens
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick Turley
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - G Bragi Walters
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Xinyi Xu
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Merete Nordentoft
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - David M Hougaard
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Thomas Werge
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Institute of Biological Psychiatry, MHC SctHans, Mental Health Services, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.
| | - Anders D Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark.
- Department of Biomedicine-Human Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
18
|
Qu L, Wang Y, Ge SN, Li N, Fu J, Zhang Y, Wang X, Jing JP, Li Y, Wang Q, Gao GD, He SM, Wang XL. Altered Activity of SK Channel Underpins Morphine Withdrawal Relevant Psychiatric Deficiency in Infralimbic to Accumbens Shell Pathway. Front Psychiatry 2019; 10:240. [PMID: 31031665 PMCID: PMC6470400 DOI: 10.3389/fpsyt.2019.00240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Drug addiction can be viewed as a chronic psychiatric disorder that is related to dysfunction of neural circuits, including reward deficits, stress surfeits, craving changes, and compromised executive function. The nucleus accumbens (NAc) plays a crucial role in regulating craving and relapse, while the medial prefrontal cortex (mPFC) represents a higher cortex projecting into the NAc that is active in the management of executive function. In this study, we investigated the role of the small conductance calcium-activated potassium channels (SK channels) in NAc and mPFC after morphine withdrawal. Action potential (AP) firing of neurons in the NAc shell was enhanced via the downregulations of the SK channels after morphine withdrawal. Furthermore, the expression of SK2 and SK3 subunits in the NAc was significantly reduced after 3 weeks of morphine withdrawal, but was not altered in the dorsal striatum. In mPFC, the SK channel subunits were differentially expressed. To be specific, the expression of SK3 was upregulated, while the expression of SK2 was unchanged. Furthermore, the AP firing in layer 5 pyramidal neurons of the infralimbic (IL) cortex was decreased via the upregulations of the SK channel-related tail current after 3 weeks of morphine withdrawal. These results suggest that the SK channel plays a specific role in reward circuits following morphine exposure and a period of drug withdrawal, making it a potential target for the prevention of relapse.
Collapse
Affiliation(s)
- Liang Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shun-Nan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jian Fu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yue Zhang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiang-Peng Jing
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shi-Ming He
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
19
|
Guo N, Zhang X, Huang M, Li X, Li Y, Zhou X, Bai J. Geranylgeranylacetone blocks the reinstatement of morphine-conditioned place preference. Neuropharmacology 2018; 143:63-70. [PMID: 30240785 DOI: 10.1016/j.neuropharm.2018.09.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 08/24/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022]
Abstract
Morphine is widely used for clinical pain management and induces the dependence. Addiction to morphine is a major public health issue. Geranylgeranylacetone (GGA) is widely used in clinic for treating ulcer. GGA induces expression of thioredoxin-1 (Trx-1) extensively. Trx-1 is a redox regulating protein and plays protecting roles in nervous system. GGA prevents mice against morphine-induced hyperlocomotion, rewarding effect, and withdrawal syndrome. However, whether GGA blocks morphine-conditioned place preference (CPP) reinstatement is still unknown. In the present study, we found that GGA administration blocked the reinstatement of morphine-CPP. The expressions of Trx-1, N-methyl d-aspartate receptor 2B subunit (NR2B), phosphorylated Ca2+/calmodulin-dependent protein kinase II (p-CaMKII), phosphorylated extracellular signaling regulated kinases (p-ERK), and phosphorylated cAMP-response element binding protein (p-CREB) were induced in nucleus accumbens (NAc) and hippocampus by morphine or GGA, whereas these proteins were not changed by morphine in GGA-treated mice. Our results indicate that GGA may prevent the reinstatement of morphine-CPP through strengthening the expression of Trx-1 and regulating NR2B/ERK pathway. Thus, we suggest that GGA may be a promising therapeutic candidate for morphine-induced relapse.
Collapse
Affiliation(s)
- Ningning Guo
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xianwen Zhang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Mengbing Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiang Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Ye Li
- Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiaoshuang Zhou
- Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical Faculty, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
20
|
Kim J, Im HI, Moon C. Intravenous morphine self-administration alters accumbal microRNA profiles in the mouse brain. Neural Regen Res 2018; 13:77-85. [PMID: 29451210 PMCID: PMC5840996 DOI: 10.4103/1673-5374.224374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A significant amount of evidence indicates that microRNAs (miRNAs) play an important role in drug addiction. The nucleus accumbens (NAc) is a critical part of the brain's reward circuit and is involved in a variety of psychiatric disorders, including depression, anxiety, and drug addiction. However, few studies have examined the expression of miRNAs and their functional roles in the NAc under conditions of morphine addiction. In this study, mice were intravenously infused with morphine (0.01, 0.03, 0.3, 1 and 3 mg/kg/infusion) and showed inverted U-shaped response. After morphine self-administration, NAc was used to analyze the functional networks of altered miRNAs and their putative target mRNAs in the NAc following intravenous self-administration of morphine. We utilized several bioinformatics tools, including Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway mapping and CyTargetLinker. We found that 62 miRNAs were altered and exhibited differential expression patterns. The putative targets were related to diverse regulatory functions, such as neurogenesis, neurodegeneration, and synaptic plasticity, as well as the pharmacological effects of morphine (receptor internalization/endocytosis). The present findings provide novel insights into the regulatory mechanisms of accumbal molecules under conditions of morphine addiction and identify several novel biomarkers associated with morphine addiction.
Collapse
Affiliation(s)
- Juhwan Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju; Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Heh-In Im
- Center for Neuroscience, Korea Institute of Science and Technology (KIST); Convergence Research Center for Diagnosis, Treatment and Care System of Dementia; Division of Biomedical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
21
|
Sultana R, Ghandi T, M. Davila A, Lee CC, Ogundele OM. Upregulated SK2 Expression and Impaired CaMKII Phosphorylation Are Shared Synaptic Defects Between 16p11.2del and 129S: Δdisc1 Mutant Mice. ASN Neuro 2018; 10:1759091418817641. [PMID: 33592687 PMCID: PMC6295693 DOI: 10.1177/1759091418817641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Ion channel gating and kinase regulation of N-methyl-D-aspartate receptor 1 activity are fundamental mechanisms that govern synaptic plasticity. In this study, we showed that two mutant models (16p11.2del and Δdisc1 ) that recapitulate aspects of human cognitive disorders shared a similar defect in N-methyl-D-aspartate receptor 1-dependent synaptic function. Our results demonstrate that the expression of small-conductance potassium channels (SK2 or KCa2.2) was significantly upregulated in the hippocampus and prefrontal cortex of 16p11.2del and 129S:Δdisc1 mutant mice. Likewise, both mutant strains exhibited an impairment of T286 phosphorylation of calcium-calmodulin-dependent kinase II (CaMKII) in the hippocampus and prefrontal cortex. In vivo neural recordings revealed that increased SK2 expression and impaired T286 phosphorylation of CaMKII coincide with a prolonged interspike interval in the hippocampal cornu ammonis-1 (CA1) field for both 16p11.2del and 129S:Δdisc1 mutant mice. These findings suggest that alteration of small conductance channels and T286 phosphorylation of CaMKII are likely shared factors underlying behavioral changes in these two genetic mouse models.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Tanya Ghandi
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Alexandra M. Davila
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, Louisiana State
University School of Veterinary Medicine, Baton Rouge, LA, USA
| |
Collapse
|
22
|
NMDA receptor dependent changes in c-fos and p-CREB signaling following extinction and reinstatement of morphine place preference. Neurosci Lett 2017; 662:147-151. [PMID: 29054430 DOI: 10.1016/j.neulet.2017.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/14/2017] [Accepted: 10/16/2017] [Indexed: 11/23/2022]
Abstract
Neural circuitry comprising the ventral tegmental area, nucleus accumbens (NAc), prefrontal cortex (PFC) and hippocampus (HIP) has a main role in reward phenomena. Previous behavioral studies indicated that intracerebroventricular administration of AP5 (NMDA glutamate receptor antagonist) and CNQX (AMPA/kainate glutamate receptor antagonist) during the extinction and before reinstatement of morphine-induced conditioned place preference (CPP) reduced the extinction period and reinstatement of morphine-CPP. Therefore, in the present study, we tried to evaluate the effect of antagonism of NMDA glutamate receptors on the p-CREB/CREB ratio and c-fos expression in the NAc, PFC and HIP during these two phases of morphine-CPP in male adult albino Wistar rats. The p-CREB/CREB ratio and c-fos levels were estimated by Western blot analysis. The results revealed that these two factors decreased by antagonism of NMDA glutamate receptors (different doses of AP5) compared to saline-control group in aforementioned regions. The reduction of molecular markers, especially the p-CREB/CREB ratio, after AP5 administration was more during the extinction period. Therefore, it can be assumed that consolidation and reconsolidation of morphine memory via intra-PFC, -NAc and -HIP NMDA glutamate receptors are in accordance with changes in p-CREB/CREB ratio and c-fos levels.
Collapse
|
23
|
Siahposht-Khachaki A, Fatahi Z, Yans A, Khodagholi F, Haghparast A. Involvement of AMPA/Kainate Glutamate Receptor in the Extinction and Reinstatement of Morphine-Induced Conditioned Place Preference: A Behavioral and Molecular Study. Cell Mol Neurobiol 2017; 37:315-328. [PMID: 27053349 PMCID: PMC11482141 DOI: 10.1007/s10571-016-0371-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Glutamate receptors in mesolimbic areas such as the nucleus accumbens, ventral tegmental area, prefrontal cortex (PFC), and hippocampus (HIP) are a component of the mechanisms of drug-induced reward and can modulate the firing pattern of dopaminergic neurons in the reward system. In addition, several lines of study have indicated that cAMP response element-binding protein (CREB) and c-fos have important role in morphine-induced conditioned place preference (CPP) induced by drugs of abuse, such as morphine, cocaine, nicotine, and alcohol. Therefore, in the present study, we investigated the changes in phosphorylated CREB (p-CREB) and c-fos induction within the nucleus accumbens (NAc), HIP, and PFC after intracerebroventricular (ICV) administration of different doses of CNQX or vehicle during extinction period or reinstatement of morphine-induced CPP. In all groups, the CPP procedure was done; afterward, the conditioning scores were recorded by Ethovision software. After behavioral test recording, we dissected out the NAc, HIP, and PFC regions and measured the p-CREB/CREB ratio and c-fos level by Western blot analysis. Our results showed that administration of CNQX significantly shortened the extinction of morphine CPP. Besides, ICV microinjection of CNQX following extinction period decreased the reinstatement of morphine CPP in extinguished rats. In molecular section, in treatment group, all mentioned factors were dose-dependently decreased in comparison with vehicle group (DMSO) after ICV microinjection of different doses of CNQX but not in pre-extinction microinjection. These findings suggested that antagonism of AMPA receptor decreased p-CREB/CREB ratio and c-fos level in the PFC, NAc, and HIP. Modulation of the drug memory reconsolidation may be useful for faster extinction of drug-induced reward and attenuation of drug-seeking behavior.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/metabolism
- Conditioning, Psychological/drug effects
- Conditioning, Psychological/physiology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Dose-Response Relationship, Drug
- Excitatory Amino Acid Antagonists/administration & dosage
- Extinction, Psychological/drug effects
- Extinction, Psychological/physiology
- Infusions, Intraventricular
- Male
- Morphine/administration & dosage
- Rats
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/metabolism
- Receptors, Kainic Acid/antagonists & inhibitors
- Receptors, Kainic Acid/metabolism
Collapse
Affiliation(s)
- Ali Siahposht-Khachaki
- Department of Physiology and Pharmacology, Mazandaran University of Medical Sciences, Ramsar International Branch, Sari, Iran
| | - Zahra Fatahi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asal Yans
- Neurobiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neurobiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran.
| |
Collapse
|
24
|
Fakira AK, Massaly N, Cohensedgh O, Berman A, Morón JA. Morphine-Associated Contextual Cues Induce Structural Plasticity in Hippocampal CA1 Pyramidal Neurons. Neuropsychopharmacology 2016; 41:2668-78. [PMID: 27170097 PMCID: PMC5026734 DOI: 10.1038/npp.2016.69] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023]
Abstract
In people with a prior history of opioid misuse, cues associated with previous drug intake can trigger relapse even after years of abstinence. Examining the processes that lead to the formation and maintenance of the memories between cues/context and the opioid may help to discover new therapeutic candidates to treat drug-seeking behavior. The hippocampus is a brain region essential for learning and memory, which has been involved in the mechanisms underlying opioid cravings. The formation of memories and associations are thought to be dependent on synaptic strengthening associated with structural plasticity of dendritic spines. Here, we assess how dendritic spines in the CA1 region of the hippocampus are affected by morphine-conditioning training. Our results show that morphine pairing with environmental cues (ie, the conditioned place preference (CPP) apparatus) triggers a significant decrease in the number of thin dendritic spines in the hippocampus. Interestingly, this effect was observed regardless of the expression of a conditioned response when mice were trained using an unpaired morphine CPP design and was absent when morphine was administered in the home cage. To investigate the mechanism underlying this structural plasticity, we examined the role of Rho GTPase in dendritic spine remodeling. We found that synaptic expression of RhoA increased with morphine conditioning and blocking RhoA signaling prevented the expression of morphine-induced CPP. Our findings uncover novel mechanisms in response to morphine-associated environmental cues and the underlying alterations in spine plasticity.
Collapse
Affiliation(s)
- Amanda K Fakira
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Nicolas Massaly
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Omid Cohensedgh
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Alexandra Berman
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Jose A Morón
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA,Department of Anesthesiology, Washington University School of Medicine, Washington University Pain Center, St Louis, MO 63110, USA, Tel: +1 314 362 0078 or +1 314 362 8565, E-mail:
| |
Collapse
|
25
|
Willis M, Trieb M, Leitner I, Wietzorrek G, Marksteiner J, Knaus HG. Small-conductance calcium-activated potassium type 2 channels (SK2, KCa2.2) in human brain. Brain Struct Funct 2016; 222:973-979. [PMID: 27357310 PMCID: PMC5334391 DOI: 10.1007/s00429-016-1258-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/20/2016] [Indexed: 12/02/2022]
Abstract
SK2 (KCa2.2) channels are voltage-independent Ca2+-activated K+ channels that regulate neuronal excitability in brain regions important for memory formation. In this study, we investigated the distribution and expression of SK2 channels in human brain by Western blot analysis and immunohistochemistry. Immunoblot analysis of human brain indicated expression of four distinct SK2 channel isoforms: the standard, the long and two short isoforms. Immunohistochemistry in paraffin-embedded post-mortem brain sections was performed in the hippocampal formation, amygdala and neocortex. In hippocampus, SK2-like immunoreactivity could be detected in strata oriens and radiatum of area CA1-CA2 and in the molecular layer. In the amygdala, SK2-like immunoreactivity was highest in the basolateral nuclei, while in neocortex, staining was mainly found enriched in layer V. Activation of SK2 channels is thought to regulate neuronal excitability in brain by contributing to the medium afterhyperpolarization. However, SK2 channels are blocked by apamin with a sensitivity that suggests heteromeric channels. The herein first shown expression of SK2 human isoform b in brain could explain the variability of electrophysiological findings observed with SK2 channels.
Collapse
Affiliation(s)
- Michael Willis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical University Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Maria Trieb
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Irmgard Leitner
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Georg Wietzorrek
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| | - Josef Marksteiner
- Department of Psychiatry and Psychotherapy A, Landeskrankenhaus Hall in Tirol, Milser Strasse 10, 6060, Hall in Tirol, Austria
| | - Hans-Günther Knaus
- Division of Molecular and Cellular Pharmacology, Medical University Innsbruck, Peter Mayr Strasse 1, 6020, Innsbruck, Austria
| |
Collapse
|
26
|
Kattalai Kailasam V, Anand P, Melyan Z. Establishing an animal model for National Acupuncture Detoxification Association (NADA) auricular acupuncture protocol. Neurosci Lett 2016; 624:29-33. [PMID: 27155456 DOI: 10.1016/j.neulet.2016.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/01/2016] [Accepted: 05/02/2016] [Indexed: 01/07/2023]
Abstract
The use of opioids in the treatment of chronic pain has increased dramatically in the past few decades making them one of the most commonly prescribed medications in the US. However, long-term use of opioids is limited by development of tolerance (decreased antinociceptive efficacy) and opioid-induced hyperalgesia - paradoxical sensitization to noxious (hyperalgesia) and non-noxious (allodynia) stimuli. Novel adjunctive therapies are needed to increase the efficacy and prolong the duration of action of opioids in chronic pain treatment. Acupuncture is often used as an adjunct therapy for the treatment of symptoms induced by non-clinical use of opioids. The National Acupuncture Detoxification Association (NADA) auricular acupuncture protocol is the most common form of acupuncture treatment for substance abuse. The standardized, easy to use and virtually painless procedure make it an attractive complementary treatment option for patients suffering from opioid-induced adverse effects. Clinical trials designed to test the efficacy of the NADA protocol yielded contradictory results. The mechanism by which NADA acupuncture could serve as a successful treatment remains unknown. Therefore, establishing an animal model of NADA acupuncture can provide a tool for investigating the efficacy and cellular mechanisms of NADA treatment. Previous studies have shown that repeated morphine administration in rodents can produce locomotor sensitization and reduce analgesic potency of a challenge dose of morphine, indicating development of morphine tolerance. Here we show that NADA acupuncture treatment can both reduce morphine-induced locomotor sensitization and prevent the development of morphine tolerance in rats, thus validating a new model for NADA acupuncture studies. Our data provides support for evidence-based use of NADA acupuncture as a new adjunctive approach that can potentially improve the side-effect profile of morphine and other prescription opioids.
Collapse
Affiliation(s)
- Vasanth Kattalai Kailasam
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA; Department of Psychiatry, Harlem Hospital Center, Columbia University Medical Center, New York, NY, USA
| | - Preeti Anand
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Zara Melyan
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
27
|
Huroy S, Kanawaty A, Magomedova L, Cummins CL, George SR, van der Kooy D, Henderson JT. EphB2 reverse signaling regulates learned opiate tolerance via hippocampal function. Behav Brain Res 2016; 300:85-96. [DOI: 10.1016/j.bbr.2015.09.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 11/27/2022]
|
28
|
In vivo activation of the SK channel in the spinal cord reduces the NMDA receptor antagonist dose needed to produce antinociception in an inflammatory pain model. Pain 2016; 156:849-858. [PMID: 25734988 DOI: 10.1097/j.pain.0000000000000124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
N-methyl-D-aspartate receptor (NMDAR) antagonists have been shown to reduce mechanical hypersensitivity in animal models of inflammatory pain. However, their clinical use is associated with significant dose-limiting side effects. Small-conductance Ca-activated K channels (SK) have been shown to modulate NMDAR activity in the brain. We demonstrate that in vivo activation of SK channels in the spinal cord can alleviate mechanical hypersensitivity in a rat model of inflammatory pain. Intrathecal (i.t.) administration of the SK channel activator, 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), attenuates complete Freund adjuvant (CFA)-induced mechanical hypersensitivity in a dose-dependent manner. Postsynaptic expression of the SK channel subunit, SK3, and apamin-sensitive SK channel-mediated currents recorded from superficial laminae are significantly reduced in the dorsal horn (DH) after CFA. Complete Freund adjuvant-induced decrease in SK-mediated currents can be reversed in vitro by bath application of NS309. In addition, immunostaining for the SK3 subunit indicates that SK3-containing channels within DH neurons can have both somatic and dendritic localization. Double immunostaining shows coexpression of SK3 and NMDAR subunit, NR1, compatible with functional interaction. Moreover, we demonstrate that i.t. coadministration of NS309 with an NMDAR antagonist reduces the dose of NMDAR antagonist, DL-2-amino-5-phosphonopentanoic acid (DL-AP5), required to produce antinociceptive effects in the CFA model. This reduction could attenuate the unwanted side effects associated with NMDAR antagonists, giving this combination potential clinical implications.
Collapse
|
29
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
30
|
Siahposht-Khachaki A, Fatahi Z, Haghparast A. Reduction of the Morphine Maintenance by Blockade of the NMDA Receptors during Extinction Period in Conditioned Place Preference Paradigm of Rats. Basic Clin Neurosci 2016; 7:341-350. [PMID: 27872695 PMCID: PMC5102563 DOI: 10.15412/j.bcn.03070407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Introduction: Activation of N-methyl-d-aspartate (NMDA) glutamate receptors in the nucleus
accumbens is a component of drug-induced reward mechanism. In addition, NMDA
receptors play a major role in brain reward system and activation of these
receptors can change firing pattern of dopamine neurons. Blockade of glutamatergic
neurotransmission reduces the expression of conditioned place preference (CPP)
induced by morphine. Therefore, in this study, by using an NMDA receptor
antagonist, DL-2-Amino-5-phosphonopentanoic acid sodium salt (AP5), the role of
NMDA receptors on the maintenance and reinstatement of morphine-CPP was
investigated. Methods: Forty-three adult male albino Wistar rats were used in this study. After
subcutaneous administration of effective dose of morphine (5 mg/kg) during CPP
paradigm, the animals received intracerebroventricular doses of AP5(1, 5, and 25
mM/5μL saline) during extinction period (free morphine stage).
Conditioning score was recorded during extinction period and reinstatement phase.
Besides, another group of the animals received a single dose administration of
AP5(5 mM) just before the administration of ineffective dose of morphine (1 mg/kg)
in reinstatement phase. Results: The results revealed that two doses of this antagonist (5 and 25 mM) significantly
shortened the extinction period of morphine-CPP but did not reduce reinstatement
induced by priming dose of morphine. Moreover, the single dose administration of
AP5(5 mM) just before prime-morphine injection decreased reinstatement of
morphine-CPP. Conclusion: These findings indicate that blockade of NMDA receptors during extinction period
reduces maintenance but not reinstatement of morphine. In addition, blocking these
receptors in reinstatement phase decreases reinstatement to extinguished
morphine.
Collapse
Affiliation(s)
- Ali Siahposht-Khachaki
- Department of Physiology and Pharmacology, School of Medicine, Ramsar International Branch, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Fatahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Rodríguez-Arias M, Aguilar MA, Miñarro J. Therapies in early development for the treatment of opiate addiction. Expert Opin Investig Drugs 2015; 24:1459-72. [PMID: 26414784 DOI: 10.1517/13543784.2015.1086746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Opiate drugs are psychoactive substances used to manage severe pain. However, their chronic use is associated with the development of addiction. Opiate addiction represents a significant public health concern. AREAS COVERED This review focuses on the most recent advances in the pharmacological treatment of opiate addiction, from those being tested in clinical trials (Phase I and II), to preclinical studies that point to new targets. Readers will gain knowledge of the wide variety of treatments used to treat opiate addiction, including their strengths and weaknesses, and the promising pharmacological targets identified by preclinical research. EXPERT OPINION Among the currently available agonist therapies, new dosage forms of buprenorphine can increase patient acceptability and compliance. New extended-release forms of naltrexone are building hope of retaining opiate-dependent subjects in a drug-free state. Unfortunately, the review of the literature shows that successful preclinical studies are often followed by discouraging results in human clinical trials. Nevertheless, all targets of potential interest should be tested exhaustively. Indeed, a number of new targets and research lines (genetics and neuroinflammation approaches) may lead to breakthroughs in the future.
Collapse
Affiliation(s)
- Marta Rodríguez-Arias
- a Universidad de Valencia, Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiologia , Avda. Blasco Ibáñez 21, 46010 Valencia, Spain +34 9 63 86 40 20 ; +34 9 63 86 46 68 ;
| | - María A Aguilar
- a Universidad de Valencia, Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiologia , Avda. Blasco Ibáñez 21, 46010 Valencia, Spain +34 9 63 86 40 20 ; +34 9 63 86 46 68 ;
| | - José Miñarro
- a Universidad de Valencia, Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiologia , Avda. Blasco Ibáñez 21, 46010 Valencia, Spain +34 9 63 86 40 20 ; +34 9 63 86 46 68 ;
| |
Collapse
|
32
|
Murthy SRK, Sherrin T, Jansen C, Nijholt I, Robles M, Dolga AM, Andreotti N, Sabatier JM, Knaus HG, Penner R, Todorovic C, Blank T. Small-conductance Ca2+-activated potassium type 2 channels regulate the formation of contextual fear memory. PLoS One 2015; 10:e0127264. [PMID: 25938421 PMCID: PMC4418695 DOI: 10.1371/journal.pone.0127264] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 04/13/2015] [Indexed: 12/28/2022] Open
Abstract
Small-conductance, Ca2+ activated K+ channels (SK channels) are expressed at high levels in brain regions responsible for learning and memory. In the current study we characterized the contribution of SK2 channels to synaptic plasticity and to different phases of hippocampal memory formation. Selective SK2 antisense-treatment facilitated basal synaptic transmission and theta-burst induced LTP in hippocampal brain slices. Using the selective SK2 antagonist Lei-Dab7 or SK2 antisense probes, we found that hippocampal SK2 channels are critical during two different time windows: 1) blockade of SK2 channels before the training impaired fear memory, whereas, 2) blockade of SK2 channels immediately after the training enhanced contextual fear memory. We provided the evidence that the post-training cleavage of the SK2 channels was responsible for the observed bidirectional effect of SK2 channel blockade on memory consolidation. Thus, Lei-Dab7-injection before training impaired the C-terminal cleavage of SK2 channels, while Lei-Dab7 given immediately after training facilitated the C-terminal cleavage. Application of the synthetic peptide comprising a leucine-zipper domain of the C-terminal fragment to Jurkat cells impaired SK2 channel-mediated currents, indicating that the endogenously cleaved fragment might exert its effects on memory formation by blocking SK2 channel-mediated currents. Our present findings suggest that SK2 channel proteins contribute to synaptic plasticity and memory not only as ion channels but also by additionally generating a SK2 C-terminal fragment, involved in both processes. The modulation of fear memory by down-regulating SK2 C-terminal cleavage might have applicability in the treatment of anxiety disorders in which fear conditioning is enhanced.
Collapse
Affiliation(s)
- Saravana R. K. Murthy
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Tessi Sherrin
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Chad Jansen
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | | | - Michael Robles
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Amalia M. Dolga
- Department of Pharmacology and Toxicology, Philipps-University Marburg, Marburg, Germany
| | - Nicolas Andreotti
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Jean-Marc Sabatier
- Laboratoire INSERM UMR1097, Parc scientifique et technologique de Luminy, Marseille, cedex 09, France
| | - Hans-Guenther Knaus
- Division for Molecular and Cellular Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Reinhold Penner
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Laboratory of Cell and Molecular Signaling, The Queen’s Medical Center, Honolulu, Hawaii, United States of America
| | - Cedomir Todorovic
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| | - Thomas Blank
- Department of Cell & Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- Institute for Neuropathology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Zhou M, Luo P, Lu Y, Li CJ, Wang DS, Lu Q, Xu XL, He Z, Guo LJ. Imbalance of HCN1 and HCN2 expression in hippocampal CA1 area impairs spatial learning and memory in rats with chronic morphine exposure. Prog Neuropsychopharmacol Biol Psychiatry 2015; 56:207-14. [PMID: 25301101 DOI: 10.1016/j.pnpbp.2014.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/13/2014] [Accepted: 09/29/2014] [Indexed: 12/12/2022]
Abstract
The hyperpolarization-activated cyclic-nucleotide-gated non-selective cation (HCN) channels play a vital role in the neurological basis underlying nervous system diseases. However, the role of HCN channels in drug addiction is not fully understood. In the present study, we investigated the expression of HCN1 and HCN2 subunits in hippocampal CA1 and the potential molecular mechanisms underlying the modulation of HCN channels in rats with chronic morphine exposure with approaches of electrophysiology, water maze, and Western blotting. We found that chronic morphine exposure (5 mg/kg, sc, for 7 days) caused an inhibition of long-term potentiation (LTP) and impairment of spatial learning and memory, which is associated with a decrease in HCN1, and an increase in HCN2 on cell membrane of hippocampal CA1 area. Additional experiments showed that the imbalance of cell membrane HCN1 and HCN2 expression under chronic morphine exposure was related to an increase in expression of TPR containing Rab8b interacting protein (TRIP8b) (1a-4) and TRIP8b (1b-2), and phosphorylation of protein kinase A (PKA) and adaptor protein 2 μ2 (AP2 μ2). Our results demonstrate the novel information that drug addiction-induced impairment of learning and memory is involved in the imbalance of HCN1 and HCN2 subunits, which is mediated by activation of TRIP8b (1a-4), TRIP8b (1b-2), PKA and AP2 μ2.
Collapse
Affiliation(s)
- Mei Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pan Luo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chang-jun Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dian-shi Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xu-lin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhi He
- Department of Neuropsychopharmacology, Medical School of China Three Gorges University, Yichang, 443002, China.
| | - Lian-jun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
34
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
35
|
Hippocampal long-term potentiation is disrupted during expression and extinction but is restored after reinstatement of morphine place preference. J Neurosci 2014; 34:527-38. [PMID: 24403152 DOI: 10.1523/jneurosci.2838-13.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Learned associations between environmental cues and morphine use play an important role in the maintenance and/or relapse of opioid addiction. Although previous studies suggest that context-dependent morphine treatment alters glutamatergic transmission and synaptic plasticity in the hippocampus, their role in morphine conditioned place preference (CPP) and reinstatement remains unknown. We investigated changes in synaptic plasticity and NMDAR expression in the hippocampus after the expression, extinction, and reinstatement of morphine CPP. Here we report that morphine CPP is associated with increased basal synaptic transmission, impaired hippocampal long-term potentiation (LTP), and increased synaptic expression of the NR1 and NR2b NMDAR subunits. Changes in synaptic plasticity, synaptic NR1 and NR2b expression, and morphine CPP were absent when morphine was not paired with a specific context. Furthermore, hippocampal LTP was impaired and synaptic NR2b expression was increased after extinction of morphine CPP, indicating that these alterations in plasticity may be involved in the mechanisms underlying the learning of drug-environment associations. After extinction of morphine CPP, a priming dose of morphine was sufficient to reinstate morphine CPP and was associated with LTP that was indistinguishable from saline control groups. In contrast, morphine CPP extinguished mice that received a saline priming dose did not show CPP and had disrupted hippocampal LTP. Finally, we found that reinstatement of morphine CPP was prevented by the selective blockade of the NR2b subunit in the hippocampus. Together, these data suggest that alterations in synaptic plasticity and glutamatergic transmission play an important role in the reinstatement of morphine CPP.
Collapse
|