1
|
Qiu Q, Yang M, Gong D, Liang H, Chen T. Potassium and calcium channels in different nerve cells act as therapeutic targets in neurological disorders. Neural Regen Res 2025; 20:1258-1276. [PMID: 38845230 DOI: 10.4103/nrr.nrr-d-23-01766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/07/2024] [Indexed: 07/31/2024] Open
Abstract
The central nervous system, information integration center of the body, is mainly composed of neurons and glial cells. The neuron is one of the most basic and important structural and functional units of the central nervous system, with sensory stimulation and excitation conduction functions. Astrocytes and microglia belong to the glial cell family, which is the main source of cytokines and represents the main defense system of the central nervous system. Nerve cells undergo neurotransmission or gliotransmission, which regulates neuronal activity via the ion channels, receptors, or transporters expressed on nerve cell membranes. Ion channels, composed of large transmembrane proteins, play crucial roles in maintaining nerve cell homeostasis. These channels are also important for control of the membrane potential and in the secretion of neurotransmitters. A variety of cellular functions and life activities, including functional regulation of the central nervous system, the generation and conduction of nerve excitation, the occurrence of receptor potential, heart pulsation, smooth muscle peristalsis, skeletal muscle contraction, and hormone secretion, are closely related to ion channels associated with passive transmembrane transport. Two types of ion channels in the central nervous system, potassium channels and calcium channels, are closely related to various neurological disorders, including Alzheimer's disease, Parkinson's disease, and epilepsy. Accordingly, various drugs that can affect these ion channels have been explored deeply to provide new directions for the treatment of these neurological disorders. In this review, we focus on the functions of potassium and calcium ion channels in different nerve cells and their involvement in neurological disorders such as Parkinson's disease, Alzheimer's disease, depression, epilepsy, autism, and rare disorders. We also describe several clinical drugs that target potassium or calcium channels in nerve cells and could be used to treat these disorders. We concluded that there are few clinical drugs that can improve the pathology these diseases by acting on potassium or calcium ions. Although a few novel ion-channel-specific modulators have been discovered, meaningful therapies have largely not yet been realized. The lack of target-specific drugs, their requirement to cross the blood-brain barrier, and their exact underlying mechanisms all need further attention. This review aims to explain the urgent problems that need research progress and provide comprehensive information aiming to arouse the research community's interest in the development of ion channel-targeting drugs and the identification of new therapeutic targets for that can increase the cure rate of nervous system diseases and reduce the occurrence of adverse reactions in other systems.
Collapse
Affiliation(s)
- Qing Qiu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Mengting Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Danfeng Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Haiying Liang
- Department of Pharmacy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Tingting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Li XT. The involvement of K + channels in depression and pharmacological effects of antidepressants on these channels. Transl Psychiatry 2024; 14:411. [PMID: 39358318 PMCID: PMC11447029 DOI: 10.1038/s41398-024-03069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
Depression is a common and complex psychiatric illness with multiple clinical symptoms, even leading to the disability and suicide. Owing to the partial understanding of the pathogenesis of depressive-like disorders, available pharmacotherapeutic strategies are developed mainly based on the "monoamine hypothesis", resulting in a limited effectiveness and a number of adverse effects in the clinical practice. The concept of multiple pathogenic factors be helpful for clarifying the etiology of depression and developing the antidepressants. It is well documented that K+ channels serve crucial roles in modulating the neuronal excitability and neurotransmitter release in the brain, and abnormality of these channels participated in the pathogenic process of diverse central nervous system (CNS) pathologies, such as seizure and Alzheimer's disease (AD). The clinical and preclinical evidence also delineates that the involvement of several types of K+ channels in depressive-like behaviors appear to be evident, suggesting these channels being one of the multiple factors in the etiology of this debilitating disorder. Emerging data manifest that diverse antidepressants impact distinct K+ channels, such as Kv, Kir and K2P, meaning the functioning of these drug via a "multi-target" manner. On the other hand, the scenario of antidepressants impinging K+ channels could render an alternative interpretation for the pharmacological effectiveness and numerous side effects in clinical trials. Furthermore, these channels serve to be considered as a "druggable target" to develop novel therapeutic compound to antagonize this psychiatry.
Collapse
Affiliation(s)
- Xian-Tao Li
- School of Medicine, Jingchu University of Technology, Jingmen, China.
- Research group of Neurological and Metabolic Disease, School of Medicine, Jingchu University of Technology, Jingmen, China.
| |
Collapse
|
3
|
Kim G, Van NTH, Nam JH, Lee W. Unraveling the Molecular Reason of Opposing Effects of α-Mangostin and Norfluoxetine on TREK-2 at the Same Binding Site. ChemMedChem 2024:e202400409. [PMID: 39145995 DOI: 10.1002/cmdc.202400409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
TWIK-related K+ channel (TREK)-2, expressed in sensory neurons, is involved in setting membrane potential, and its modulations contributes to the generation of nociceptive signals. Although acute and chronic pain is a common symptom experienced by patients with various conditions, most existing analgesics exhibit low efficacy and are associated with adverse effects. For this reason, finding the novel modulator of TREK-2 is of significance for the development of new analgesics. Recent studies have shown that α-Mangostin (α-MG) activates TREK-2, facilitating analgesic effects, yet the underlying molecular mechanisms remain elusive. Intriguingly, even though norfluoxetine (NFx) is known to inhibit TREK-2, α-MG is also observed to share a same binding site with NFx, and this implies that TREK-2 might be modulated in a highly complicated manner. Therefore, we examine the mechanism of how TREK-2 is activated by α-MG using computational methods and patch clamp experiments in the present study. Based on these results, we offer an explanation of how α-MG and NFx exhibit opposing effects at the same binding site of TREK-2. These findings will broaden our understanding of TREK-2 modulation, providing clues for designing novel analgesic drugs.
Collapse
Affiliation(s)
- Gangrae Kim
- Department of Biochemistry, Kangwon National University, College of Natural Sciences, Chuncheon, 24341, Republic of Korea
| | - Nhung Thi Hong Van
- Department of Physiology, Dongguk University, College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University, College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Wook Lee
- Department of Biochemistry, Kangwon National University, College of Natural Sciences, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
4
|
Du C, Chen L, Liu G, Yuan F, Zhang Z, Rong M, Mo G, Liu C. Tick-Derived Peptide Blocks Potassium Channel TREK-1. Int J Mol Sci 2024; 25:8377. [PMID: 39125945 PMCID: PMC11312834 DOI: 10.3390/ijms25158377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Ticks transmit a variety of pathogens, including rickettsia and viruses, when they feed on blood, afflicting humans and other animals. Bioactive components acting on inflammation, coagulation, and the immune system were reported to facilitate ticks' ability to suck blood and transmit tick-borne diseases. In this study, a novel peptide, IstTx, from an Ixodes scapularis cDNA library was analyzed. The peptide IstTx, obtained by recombinant expression and purification, selectively inhibited a potassium channel, TREK-1, in a dose-dependent manner, with an IC50 of 23.46 ± 0.22 μM. The peptide IstTx exhibited different characteristics from fluoxetine, and the possible interaction of the peptide IstTx binding to the channel was explored by molecular docking. Notably, extracellular acidification raised its inhibitory efficacy on the TREK-1 channel. Our results found that the tick-derived peptide IstTx blocked the TREK-1 channel and provided a novel tool acting on the potassium channel.
Collapse
Affiliation(s)
- Canwei Du
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Linyan Chen
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Guohao Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Fuchu Yuan
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410006, China
| | - Zheyang Zhang
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Mingqiang Rong
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410006, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, No. 387-201 Heming Street, Chengdu 610212, China
| | - Guoxiang Mo
- College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Changjun Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| |
Collapse
|
5
|
Deal PE, Lee H, Mondal A, Lolicato M, Mendonça PRFD, Black H, Jang S, El-Hilali X, Bryant C, Isacoff EY, Renslo AR, Minor DL. Development of covalent chemogenetic K 2P channel activators. Cell Chem Biol 2024; 31:1305-1323.e9. [PMID: 39029456 PMCID: PMC11433823 DOI: 10.1016/j.chembiol.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/19/2024] [Accepted: 06/19/2024] [Indexed: 07/21/2024]
Abstract
K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (covalent activation of TREK family K+ channels to clamp membrane potential) that leverages the discovery of a K2P modulator pocket site that reacts with electrophile-bearing derivatives of a TREK subfamily small-molecule activator, ML335, to activate the channel irreversibly. We show that CATKLAMP can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a means to alter K2P channel activity that should facilitate molecular and systems level studies of K2P function and enable the search for new K2P modulators.
Collapse
Affiliation(s)
- Parker E Deal
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | | | - Holly Black
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Xochina El-Hilali
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Clifford Bryant
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Weill Neurohub, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 93858-2330, USA.
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 93858-2330, USA; California Institute for Quantitative Biomedical Research, University of California, San Francisco, San Francisco, CA 93858-2330, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 93858-2330, USA.
| |
Collapse
|
6
|
Wen W, Zhou J, Zhan C, Wang J. Microglia as a Game Changer in Epilepsy Comorbid Depression. Mol Neurobiol 2024; 61:4021-4037. [PMID: 38048030 DOI: 10.1007/s12035-023-03810-0] [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: 12/26/2022] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
As one of the most common neurological diseases, epilepsy is often accompanied by psychiatric disorders. Depression is the most universal comorbidity of epilepsy, especially in temporal lobe epilepsy (TLE). Therefore, it is urgently needed to figure out potential mechanisms and the optimization of therapeutic strategies. Microglia play a pivotal role in the coexistent relationship between epilepsy and depression. Activated microglia released cytokines like IL-6 and IL-1β, orchestrating neuroinflammation especially in the hippocampus, worsening both depression and epilepsy. The decrease of intracellular K+ is a common part in various molecular changes. The P2X7-NLRP3-IL-1β is a major inflammatory pathway that disrupts brain network. Extra ATP and CX3CL1 also lead to neuronal excitotoxicity and blood-brain barrier (BBB) disruption. Regulating neuroinflammation aiming at microglia-related molecules is capable of suspending the vicious mutual aggravating circle of epilepsy and depression. Other overlaps between epilepsy and depression lie in transcriptomic, neuroimaging, diagnosis and treatment. Hippocampal sclerosis (HS) and amygdala enlargement (AE) may be the underlying macroscopic pathological changes according to current studies. Extant evidence shows that cognitive behavioral therapy (CBT) and antidepressants like selective serotonin-reuptake inhibitors (SSRIs) are safe, but the effect is limited. Improvement in depression is likely to reduce the frequency of seizure. More comprehensive experiments are warranted to better understand the relationship between them.
Collapse
Affiliation(s)
- Wenrong Wen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Avenue North, Guangzhou, 1838, Guangdong Province, China
- The First Clinical Medicine College, Southern Medical University, Guangzhou, Guangdong Province, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jingsheng Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Avenue North, Guangzhou, 1838, Guangdong Province, China
- The First Clinical Medicine College, Southern Medical University, Guangzhou, Guangdong Province, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chang'an Zhan
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jun Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou Avenue North, Guangzhou, 1838, Guangdong Province, China.
- The First Clinical Medicine College, Southern Medical University, Guangzhou, Guangdong Province, China.
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
7
|
Zhou X, Zhao C, Xu H, Xu Y, Zhan L, Wang P, He J, Lu T, Gu Y, Yang Y, Xu C, Chen Y, Liu Y, Zeng Y, Tian F, Chen Q, Xie X, Liu J, Hu H, Li J, Zheng Y, Guo J, Gao Z. Pharmacological inhibition of Kir4.1 evokes rapid-onset antidepressant responses. Nat Chem Biol 2024; 20:857-866. [PMID: 38355723 DOI: 10.1038/s41589-024-01555-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
Major depressive disorder, a prevalent and severe psychiatric condition, necessitates development of new and fast-acting antidepressants. Genetic suppression of astrocytic inwardly rectifying potassium channel 4.1 (Kir4.1) in the lateral habenula ameliorates depression-like phenotypes in mice. However, Kir4.1 remains an elusive drug target for depression. Here, we discovered a series of Kir4.1 inhibitors through high-throughput screening. Lys05, the most potent one thus far, effectively suppressed native Kir4.1 channels while displaying high selectivity against established targets for rapid-onset antidepressants. Cryogenic-electron microscopy structures combined with electrophysiological characterizations revealed Lys05 directly binds in the central cavity of Kir4.1. Notably, a single dose of Lys05 reversed the Kir4.1-driven depression-like phenotype and exerted rapid-onset (as early as 1 hour) antidepressant actions in multiple canonical depression rodent models with efficacy comparable to that of (S)-ketamine. Overall, we provided a proof of concept that Kir4.1 is a promising target for rapid-onset antidepressant effects.
Collapse
Affiliation(s)
- Xiaoyu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- College of Pharmacy, University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Zhao
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyan Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yixiang Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Li Zhan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Pei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingyi He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Taotao Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yueling Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Yang
- Liangzhu Laboratory, Zhejiang University School of Medicine, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou, China
| | - Chanjuan Xu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyang Chen
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxuan Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fuyun Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China
| | - Qian Chen
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jianfeng Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hailan Hu
- Liangzhu Laboratory, Zhejiang University School of Medicine, New Cornerstone Science Laboratory, Zhejiang University, Hangzhou, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yueming Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Jiangtao Guo
- Department of Biophysics and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- College of Pharmacy, University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
- School of Pharmacy, Henan University, Kaifeng, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
| |
Collapse
|
8
|
Roy-Chowdhury S, Jang S, Abderemane-Ali F, Naughton F, Grabe M, Minor DL. Structure of the human K 2P13.1(THIK-1) channel reveals a novel hydrophilic pore restriction and lipid cofactor site. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600491. [PMID: 38979306 PMCID: PMC11230452 DOI: 10.1101/2024.06.26.600491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The halothane-inhibited K2P leak potassium channel K2P13.1 (THIK-1)1-3 is found in diverse cells1,4 including neurons1,5 and microglia6-8 where it affects surveillance6, synaptic pruning7, phagocytosis7, and inflammasome-mediated interleukin-1β release6,8,9. As with many K2Ps1,5,10-14 and other voltage-gated ion channel (VGIC) superfamily members3,15,16, polyunsaturated fatty acid (PUFA) lipids modulate K2P13.1 (THIK-1)1,5,14,17 via a poorly understood mechanism. Here, we present cryo-electronmicroscopy (cryo-EM) structures of human K2P13.1 (THIK-1) and mutants in lipid nanodiscs and detergent. These reveal that, unlike other K2Ps13,18-24, K2P13.1 (THIK-1) has a two-chamber aqueous inner cavity obstructed by a M4 transmembrane helix tyrosine (Tyr273, the flow restrictor). This hydrophilic barrier can be opened by an activatory mutation, S136P25, at natural break in the M2 transmembrane helix and by intrinsic channel dynamics. The structures also reveal a buried lipid in the P1/M4 intersubunit interface at a location, the PUFA site, that coincides with the TREK subfamily K2P modulator pocket for small molecule agonists18,26,27. This overlap, together with the effects of mutation on K2P13.1 (THIK-1) PUFA responses, indicates that the PUFA site lipids are K2P13.1 (THIK-1) cofactors. Comparison with the PUFA-responsive VGIC Kv7.1 (KCNQ1)28-31 reveals a shared role for the equivalent pore domain intersubunit interface in lipid modulation, providing a framework for dissecting the effects of PUFAs on the VGIC superfamily. Our findings reveal the unique architecture underlying K2P13.1 (THIK-1) function, highlight the importance of the P1/M4 interface in control of K2Ps by both natural and synthetic agents, and should aid development of THIK subfamily modulators for diseases such as neuroinflammation6,32 and autism6.
Collapse
Affiliation(s)
- Shatabdi Roy-Chowdhury
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Seil Jang
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Fiona Naughton
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Michael Grabe
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
| | - Daniel L. Minor
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, California 93858-2330 USA
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| |
Collapse
|
9
|
Okada M, Tran TTT. Effect of chronic administration of ostruthin on depression-like behavior in chronically stressed mice. IBRO Neurosci Rep 2024; 16:622-628. [PMID: 38832088 PMCID: PMC11144753 DOI: 10.1016/j.ibneur.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
We have previously shown that a single dose of a TREK-1 channel activator, ostruthin, exhibited antidepressant and anxiolytic effects in acute behavioral test models in mice. To assess the potential clinical application, it is essential to evaluate the effects of long-term administration of ostruthin in a chronically stressed mouse model, which is considered to be similar to the clinical condition of major depression in humans. Here, we tested the effects of a single and a 7-day administration of ostruthin on mice that were subjected to chronic unpredictable mild stress (CUMS). A single administration of ostruthin showed antidepressive effects in the tail suspension and forced swim tests of CUMS-treated mice. Unexpectedly, the 7-day administration exhibited only insignificant antidepressive and anxiolytic effects. The 7-day regimen did not affect food intake or body-weight gain, suggesting the absence of apparent cytotoxicity. The mice receiving the 7-day administration had significantly lower blood concentrations of ostruthin compared to those receiving a single dose, suggesting an upregulation of drug-metabolizing activities. These findings suggest that there is a need for stable TREK-1 channel activators that are not affected by drug metabolism.
Collapse
Affiliation(s)
- Masayoshi Okada
- Department of Medical LifeScience, College of Life Science, Kurashiki University of Science and the Arts, Kurashiki, Okayama 712-8505, Japan
| | - Thi Thu Thuy Tran
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, Hanoi, Viet Nam
| |
Collapse
|
10
|
Francis-Oliveira J, Higa GSV, Viana FJC, Cruvinel E, Carlos-Lima E, da Silva Borges F, Zampieri TT, Rebello FP, Ulrich H, De Pasquale R. TREK-1 inhibition promotes synaptic plasticity in the prelimbic cortex. Exp Neurol 2024; 373:114652. [PMID: 38103709 DOI: 10.1016/j.expneurol.2023.114652] [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: 10/06/2023] [Revised: 11/28/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Synaptic plasticity is one of the putative mechanisms involved in the maturation of the prefrontal cortex (PFC) during postnatal development. Early life stress (ELS) affects the shaping of cortical circuitries through impairment of synaptic plasticity supporting the onset of mood disorders. Growing evidence suggests that dysfunctional postnatal maturation of the prelimbic division (PL) of the PFC might be related to the emergence of depression. The potassium channel TREK-1 has attracted particular interest among many factors that modulate plasticity, concerning synaptic modifications that could underlie mood disorders. Studies have found that ablation of TREK-1 increases the resilience to depression, while rats exposed to ELS exhibit higher TREK-1 levels in the PL. TREK-1 is regulated by multiple intracellular transduction pathways including the ones activated by metabotropic receptors. In the hippocampal neurons, TREK-1 interacts with the serotonergic system, one of the main factors involved in the action of antidepressants. To investigate possible mechanisms related to the antidepressant role of TREK-1, we used brain slice electrophysiology to evaluate the effects of TREK-1 pharmacological blockade on synaptic plasticity at PL circuitry. We extended this investigation to animals subjected to ELS. Our findings suggest that in non-stressed animals, TREK-1 activity is required for the reduction of synaptic responses mediated by the 5HT1A receptor activation. Furthermore, we demonstrate that TREK-1 blockade promotes activity-dependent long-term depression (LTD) when acting in synergy with 5HT1A receptor stimulation. On the other hand, in ELS animals, TREK-1 blockade reduces synaptic transmission and facilitates LTD expression. These results indicate that TREK-1 inhibition stimulates synaptic plasticity in the PL and this effect is more pronounced in animals subjected to ELS during postnatal development.
Collapse
Affiliation(s)
- José Francis-Oliveira
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil; Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP 09210-580, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Estevão Carlos-Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernando da Silva Borges
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Thais Tessari Zampieri
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Fernanda Pereira Rebello
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, SP 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, SP 05508-000, Brazil.
| |
Collapse
|
11
|
Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW, Xue M. Ion Channel Dysregulation Following Intracerebral Hemorrhage. Neurosci Bull 2024; 40:401-414. [PMID: 37755675 PMCID: PMC10912428 DOI: 10.1007/s12264-023-01118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/14/2023] [Indexed: 09/28/2023] Open
Abstract
Injury to the brain after intracerebral hemorrhage (ICH) results from numerous complex cellular mechanisms. At present, effective therapy for ICH is limited and a better understanding of the mechanisms of brain injury is necessary to improve prognosis. There is increasing evidence that ion channel dysregulation occurs at multiple stages in primary and secondary brain injury following ICH. Ion channels such as TWIK-related K+ channel 1, sulfonylurea 1 transient receptor potential melastatin 4 and glutamate-gated channels affect ion homeostasis in ICH. They in turn participate in the formation of brain edema, disruption of the blood-brain barrier, and the generation of neurotoxicity. In this review, we summarize the interaction between ions and ion channels, the effects of ion channel dysregulation, and we discuss some therapeutics based on ion-channel modulation following ICH.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Qiuyang Su
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
12
|
Coppola T, Daziano G, Legroux I, Béraud-Dufour S, Blondeau N, Lebrun P. Unlocking Therapeutic Synergy: Tailoring Drugs for Comorbidities such as Depression and Diabetes through Identical Molecular Targets in Different Cell Types. Cells 2023; 12:2768. [PMID: 38067196 PMCID: PMC10706795 DOI: 10.3390/cells12232768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Research in the field of pharmacology aims to generate new treatments for pathologies. Nowadays, there are an increased number of chronic disorders that severely and durably handicap many patients. Among the most widespread pathologies, obesity, which is often associated with diabetes, is constantly increasing in incidence, and in parallel, neurodegenerative and mood disorders are increasingly affecting many people. For years, these pathologies have been so frequently observed in the population in a concomitant way that they are considered as comorbidities. In fact, common mechanisms are certainly at work in the etiology of these pathologies. The main purpose of this review is to show the value of anticipating the effect of baseline treatment of a condition on its comorbidity in order to obtain concomitant positive actions. One of the implications would be that by understanding and targeting shared molecular mechanisms underlying these conditions, it may be possible to tailor drugs that address both simultaneously. To this end, we firstly remind readers of the close link existing between depression and diabetes and secondly address the potential benefit of the pleiotropic actions of two major active molecules used to treat central and peripheral disorders, first a serotonin reuptake inhibitor (Prozac ®) and then GLP-1R agonists. In the second part, by discussing the therapeutic potential of new experimental antidepressant molecules, we will support the concept that a better understanding of the intracellular signaling pathways targeted by pharmacological agents could lead to future synergistic treatments targeting solely positive effects for comorbidities.
Collapse
Affiliation(s)
- Thierry Coppola
- CNRS, IPMC, Université Côte d’Azur, Sophia Antipolis, F-06560 Valbonne, France; (G.D.); (I.L.); (S.B.-D.); (N.B.)
| | | | | | | | | | - Patricia Lebrun
- CNRS, IPMC, Université Côte d’Azur, Sophia Antipolis, F-06560 Valbonne, France; (G.D.); (I.L.); (S.B.-D.); (N.B.)
| |
Collapse
|
13
|
Deal PE, Lee H, Mondal A, Lolicato M, de Mendonca PRF, Black H, El-Hilali X, Bryant C, Isacoff EY, Renslo AR, Minor DL. Development of covalent chemogenetic K 2P channel activators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.15.561774. [PMID: 37905049 PMCID: PMC10614804 DOI: 10.1101/2023.10.15.561774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
K2P potassium channels regulate excitability by affecting cellular resting membrane potential in the brain, cardiovascular system, immune cells, and sensory organs. Despite their important roles in anesthesia, arrhythmia, pain, hypertension, sleep, and migraine, the ability to control K2P function remains limited. Here, we describe a chemogenetic strategy termed CATKLAMP (Covalent Activation of TREK family K+ channels to cLAmp Membrane Potential) that leverages the discovery of a site in the K2P modulator pocket that reacts with electrophile-bearing derivatives of a TREK subfamily small molecule activator, ML335, to activate the channel irreversibly. We show that the CATKLAMP strategy can be used to probe fundamental aspects of K2P function, as a switch to silence neuronal firing, and is applicable to all TREK subfamily members. Together, our findings exemplify a new means to alter K2P channel activity that should facilitate studies both molecular and systems level studies of K2P function and enable the search for new K2P modulators.
Collapse
Affiliation(s)
- Parker E. Deal
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Abhisek Mondal
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
| | | | - Holly Black
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Xochina El-Hilali
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Clifford Bryant
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Ehud Y. Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California 94720, United States
- Weill Neurohub, University of California, Berkeley, Berkeley, California 94720, United States
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 93858-2330 USA
| | - Daniel L. Minor
- Cardiovascular Research Institute, University of California, San Francisco, California 93858-2330 USA
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 USA
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, California 93858-2330 USA
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 93858-2330 USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 93858-2330 USA
| |
Collapse
|
14
|
Lee EH, Park JE, Gotina L, Han YE, Viswanath ANI, Yoo S, Moon B, Hwang JY, Park WK, Cho Y, Song C, Min SJ, Hwang EM, Lee H, Pae AN, Roh EJ, Oh SJ. Novel potent blockers for TWIK-1/TREK-1 heterodimers as potential antidepressants. Biomed Pharmacother 2023; 165:115139. [PMID: 37454597 DOI: 10.1016/j.biopha.2023.115139] [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: 05/09/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
TREK-1 (TWIK-related potassium channel-1) is a subunit of the two-pore domain potassium (K2p) channel and is widely expressed in the brain. TREK-1 knockout mice were shown to have antidepressant-like effects, providing evidence for the channel's potential as a therapeutic target. However, currently there is no good pharmacological inhibitor specifically targeting TREK-1 containing K2p channels that also displays similar antidepressant-like effects. Here, we sought to find selective and potent inhibitors for TREK-1 related dimers both in vitro and in vivo. We synthesized and evaluated 2-hydroxy-3-phenoxypropyl piperidine derivatives yielding a library from which many TREK-1 targeting candidates emerged. Among these, hydroxyl-phenyl- (2a), piperidino- (2g), and pyrrolidino- (2h) piperidinyl substituted compounds showed high potencies to TREK-1 homodimers with significant antidepressant-like effects in forced swim test and tail suspension test. Interestingly, these compounds were found to have high potencies to TWIK-1/TREK-1 heterodimers. Contrastingly, difluoropiperidinyl-4-fluorophenoxy (3e) and 4-hydroxyphenyl-piperidinyl-4-fluorophenoxy (3j) compounds had high potencies to TREK-1 homodimer but lower potency to TWIK-1/TREK-1 heterodimers without significant antidepressant-like effects. We observed positive correlation between inhibition potency to TWIK-1/TREK-1 and immobility time, and no correlation between inhibition potency to TREK-1 homodimer and immobility time. This was consistent with molecular docking simulations of selected compounds to TREK-1 homodimeric and TWIK-1/TREK-1 heterodimeric models. Existing antidepressant fluoxetine was also found to potently inhibit TWIK-1/TREK-1 heterodimers. Our study reveals novel potent TWIK-1/TREK-1 inhibitors 2a, 2g, and 2h as potential antidepressants and suggest that the TWIK-1/TREK-1 heterodimer could be a potential novel molecular therapeutic target for antidepressants.
Collapse
Affiliation(s)
- Elliot H Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jung-Eun Park
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Chemistry, Sogang University, Baekbeomno 35, Mapo-gu, Seoul, Republic of Korea
| | - Lizaveta Gotina
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Ambily Nath Indu Viswanath
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Seonguk Yoo
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bongjin Moon
- Department of Chemistry, Sogang University, Baekbeomno 35, Mapo-gu, Seoul, Republic of Korea
| | - Jin-Young Hwang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Woo Kyu Park
- Rare Disease Therapeutic Technology Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yoonjeong Cho
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea; Biomedical Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sun-Joon Min
- Department of Chemical & Molecular Engineering/Applied Chemistry, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Hyunbeom Lee
- Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of HY-KIST Bio-convergence, Hanyang University, Seoul 04763, Republic of Korea
| | - Ae Nim Pae
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
15
|
Sha Z, Xu J, Li N, Li O. Regulatory Molecules of Synaptic Plasticity in Anxiety Disorder. Int J Gen Med 2023; 16:2877-2886. [PMID: 37435365 PMCID: PMC10332425 DOI: 10.2147/ijgm.s413176] [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: 03/19/2023] [Accepted: 06/19/2023] [Indexed: 07/13/2023] Open
Abstract
Synaptic plasticity is the capacity of synaptic transmission between neurons to be strengthened or weakened. There are many signal molecules accumulated in the presynaptic and postsynaptic membranes that can lead to the regulation of synaptic plasticity and involvement in numerous of neurological and psychiatric diseases, including anxiety disorder. However, the regulatory mechanisms of synaptic plasticity in the development of anxiety disorder have not been well summarized. This review mainly aims to discuss the biological functions and mechanisms of synaptic plasticity-related molecules in anxiety disorder, with a particular focus on the metabotropic glutamate receptors, brain-derived neurotrophic factor, hyperpolarization-activated cyclic nucleotide-gated channels, and postsynaptic density 95. The summarized functions and mechanisms of synaptic plasticity-related molecules in anxiety will provide insight into novel neuroplasticity modifications for targeted therapy for anxiety.
Collapse
Affiliation(s)
- Zhongwei Sha
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Xu
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Nana Li
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Ou Li
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
16
|
Zhang Y, Fu J, Han Y, Feng D, Yue S, Zhou Y, Luo Z. Two-Pore-Domain Potassium Channel TREK-1 Mediates Pulmonary Fibrosis through Macrophage M2 Polarization and by Direct Promotion of Fibroblast Differentiation. Biomedicines 2023; 11:biomedicines11051279. [PMID: 37238950 DOI: 10.3390/biomedicines11051279] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by myofibroblast proliferation and abnormal accumulation of extracellular matrix in the lungs. After lung injury, M2 macrophages mediate the pathogenesis of pulmonary fibrosis by secreting fibrotic cytokines that promote myofibroblast activation. The TWIK-related potassium channel (TREK-1, also known as KCNK2) is a K2P channel that is highly expressed in cardiac, lung, and other tissues; it worsens various tumors, such as ovarian cancer and prostate cancer, and mediates cardiac fibrosis. However, the role of TREK-1 in lung fibrosis remains unclear. This study aimed to examine the effects of TREK-1 on bleomycin (BLM)-induced lung fibrosis. The results show that TREK-1 knockdown, mediated by the adenovirus or pharmacological inhibition of TREK-1 with fluoxetine, resulted in diminished BLM-induced lung fibrosis. TREK-1 overexpression in macrophages remarkably increased the M2 phenotype, resulting in fibroblast activation. Furthermore, TREK-1 knockdown and fluoxetine administration directly reduced the differentiation of fibroblasts to myofibroblasts by inhibiting the focal adhesion kinase (FAK)/p38 mitogen-activated protein kinases (p38)/Yes-associated protein (YAP) signaling pathway. In conclusion, TREK-1 plays a central role in the pathogenesis of BLM-induced lung fibrosis, which serves as a theoretical basis for the inhibition of TREK-1 as a potential therapy protocol for lung fibrosis.
Collapse
Affiliation(s)
- Yunna Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jiafeng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Yang Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Shaojie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, China
| |
Collapse
|
17
|
Schroeter CB, Nelke C, Schewe M, Spohler L, Herrmann AM, Müntefering T, Huntemann N, Kuzikov M, Gribbon P, Albrecht S, Bock S, Hundehege P, Neelsen LC, Baukrowitz T, Seebohm G, Wünsch B, Bittner S, Ruck T, Budde T, Meuth SG. Validation of TREK1 ion channel activators as an immunomodulatory and neuroprotective strategy in neuroinflammation. Biol Chem 2023; 404:355-375. [PMID: 36774650 DOI: 10.1515/hsz-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/16/2023] [Indexed: 02/13/2023]
Abstract
Modulation of two-pore domain potassium (K2P) channels has emerged as a novel field of therapeutic strategies as they may regulate immune cell activation and metabolism, inflammatory signals, or barrier integrity. One of these ion channels is the TWIK-related potassium channel 1 (TREK1). In the current study, we report the identification and validation of new TREK1 activators. Firstly, we used a modified potassium ion channel assay to perform high-throughput-screening of new TREK1 activators. Dose-response studies helped to identify compounds with a high separation between effectiveness and toxicity. Inside-out patch-clamp measurements of Xenopus laevis oocytes expressing TREK1 were used for further validation of these activators regarding specificity and activity. These approaches yielded three substances, E1, B3 and A2 that robustly activate TREK1. Functionally, we demonstrated that these compounds reduce levels of adhesion molecules on primary human brain and muscle endothelial cells without affecting cell viability. Finally, we studied compound A2 via voltage-clamp recordings as this activator displayed the strongest effect on adhesion molecules. Interestingly, A2 lacked TREK1 activation in the tested neuronal cell type. Taken together, this study provides data on novel TREK1 activators that might be employed to pharmacologically modulate TREK1 activity.
Collapse
Affiliation(s)
- Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Marcus Schewe
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Lucas Spohler
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Alexander M Herrmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Thomas Müntefering
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune mediated diseases (CIMD), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune mediated diseases (CIMD), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Sarah Albrecht
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Stefanie Bock
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Petra Hundehege
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Lea Christine Neelsen
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Guiscard Seebohm
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Robert-Koch-Straße 27A, D-48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
18
|
Blocking Two-Pore Domain Potassium Channel TREK-1 Inhibits the Activation of A1-Like Reactive Astrocyte Through the NF-κB Signaling Pathway in a Rat Model of Major Depressive Disorder. Neurochem Res 2023; 48:1737-1754. [PMID: 36670238 PMCID: PMC10119044 DOI: 10.1007/s11064-023-03857-4] [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: 10/01/2022] [Revised: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/22/2023]
Abstract
Major depressive disorder (MDD) refers to a widespread psychiatric disorder. Astrocytes play a pivotal role in regulating inflammation which is a well-acknowledged key component in depression pathogenesis. However, the effects of the neuroinflammation-inducing A1-like astrocytes on MDD are still unknown. TWIK-related K+ channel 1 (TREK-1) has been demonstrated to regulate the action of antidepressants. Nevertheless, its mechanisms and effects on A1-like astrocyte stimulation in MDD are not clear. Therefore, we conducted in vivo and in vitro experiments using TREK-1 specific inhibitor spadin. In vivo, rats were subjected to a 6-week chronic unpredictable mild stress (CUMS) followed by spadin treatment. Behavioral tests were employed to surveil depressive-like behaviors. Hippocampal proteomic analysis was carried out with the purpose of identifying differentially expressed proteins after CUMS and spadin treatments. In vitro, astrocyte-conditioned medium and spadin were used to treat rat astrocyte cell line. The activated microglia, inflammatory factors, A1 astrocyte markers, and activated nuclear factor kappa B (NF-κB) pathway were later analyzed using immunofluorescence, western blot, and RT-qPCR. Our findings indicated that blockage of TREK-1 reduced CUMS-induced depressive-like behavior in rats, inhibited the microglial stimulation, reduced inflammatory factor levels, and suppressed the activation of A1-like reactive astrocytes in the hippocampus. We also verified that the suppression of A1-like astrocytes by spadin necessitated the NF-κB pathway. According to the findings, blocking TREK-1 inhibited the activation of A1-like reactive astrocytes via the NF-κB signaling pathway in MDD. Our study preliminarily identifies a novel antidepressant mechanism of TREK-1 action and provides a therapeutic path for MDD.
Collapse
|
19
|
Hu G, Zhang M, Wang Y, Yu M, Zhou Y. Potential of Heterogeneous Compounds as Antidepressants: A Narrative Review. Int J Mol Sci 2022; 23:ijms232213776. [PMID: 36430254 PMCID: PMC9692659 DOI: 10.3390/ijms232213776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Depression is a globally widespread disorder caused by a complicated interplay of social, psychological, and biological factors. Approximately 280 million people are suffering from depression worldwide. Traditional frontline antidepressants targeting monoamine neurotransmitters show unsatisfactory effects. The development and application of novel antidepressants for dissimilar targets are on the agenda. This review characterizes the antidepressant effects of multiple endogenous compounds and/or their targets to provide new insight into the working mechanism of antidepressants. We also discuss perspectives and challenges for the generation of novel antidepressants.
Collapse
Affiliation(s)
- Gonghui Hu
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao 266071, China
| | - Meng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao 266071, China
| | - Yuyang Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Ming Yu
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao 266000, China
| | - Yu Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao 266071, China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao 266000, China
- Correspondence:
| |
Collapse
|
20
|
Xu H, Ding Y, Qi X, Zhang ZJ, Su J. Ameliorated Neurogenesis Deficits in Dentate Gyrus May Underly the Pronounced Antidepressant Effect of TREK-1 Potassium Channel Blockade in Rats with Depressive-like Behavior. ACS Chem Neurosci 2022; 13:3068-3077. [PMID: 36269040 DOI: 10.1021/acschemneuro.2c00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Depression is considered to be the most common mental disorder and is probed by several studies that chronic mild stress contributes to depression, and fortunately, most antidepressants ameliorate depressive-like behavior accompanied with reversed hippocampal neurogenesis defects. In our present study, we confirmed that different antidepressants repaired the stress-induced neuronal and behavioral deficits by modulating adult hippocampal neurogenesis. Antidepressant treatment restored the adult hippocampal neurodegeneration, which was impaired by chronic unpredicted mild stress displaying decreased proliferation and neuronal differentiation but increased apoptosis of newly formed neurons in dentate gyrus. Notably, sucrose preference ratio significantly correlated with both neuronal differentiation proportion and newborn apoptosis proportion, suggesting a mechanistic relationship between neurogenesis and behavior. Indeed, the neotype TREK-1 potassium channel blocker expressed an earlier and pronounced antidepressant manifestation compared to the traditional selective serotonin-reuptake inhibitors fluoxetine. We therefore conclude that the administration of TREK-1 potassium channel antagonism can reverse the depressive deficits caused by chronic stress quickly via regulation of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Hua Xu
- Department of Neurology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou, Jiangsu 213200, China.,Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yingpeng Ding
- Department of Cardiology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou 213200, Jiangsu, China
| | - Xinyang Qi
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.,The Brain Cognition and Brain Disease Institute (BCBDI), CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jianhua Su
- Department of Neurology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou, Jiangsu 213200, China
| |
Collapse
|
21
|
Kim SS, Bae Y, Kwon O, Kwon SH, Seo JB, Hwang EM, Park JY. β-COP Regulates TWIK1/TREK1 Heterodimeric Channel-Mediated Passive Conductance in Astrocytes. Cells 2022; 11:cells11203322. [PMID: 36291187 PMCID: PMC9600989 DOI: 10.3390/cells11203322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Mature astrocytes are characterized by a K+ conductance (passive conductance) that changes with a constant slope with voltage, which is involved in K+ homeostasis in the brain. Recently, we reported that the tandem of pore domains in a weak inward rectifying K+ channel (TWIK1 or KCNK1) and TWIK-related K+ channel 1 (TREK1 or KCNK2) form heterodimeric channels that mediate passive conductance in astrocytes. However, little is known about the binding proteins that regulate the function of the TWIK1/TREK1 heterodimeric channels. Here, we found that β-coat protein (COP) regulated the surface expression and activity of the TWIK1/TREK1 heterodimeric channels in astrocytes. β-COP binds directly to TREK1 but not TWIK1 in a heterologous expression system. However, β-COP also interacts with the TWIK1/TREK1 heterodimeric channel in a TREK1 dependent manner and enhances the surface expression of the heterodimeric channel in astrocytes. Consequently, it regulates TWIK1/TREK1 heterodimeric channel-mediated passive conductance in astrocytes in the mouse brain. Taken together, these results suggest that β-COP is a potential regulator of astrocytic passive conductance in the brain.
Collapse
Affiliation(s)
- Seong-Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Osung Kwon
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Seung-Hae Kwon
- Seoul Center, Korea Basic Science Institute (KBSI), Seoul 02841, Korea
| | - Jong Bok Seo
- Seoul Center, Korea Basic Science Institute (KBSI), Seoul 02841, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-3290-5637
| |
Collapse
|
22
|
Herrera-Pérez S, Rueda-Ruzafa L, Campos-Ríos A, Fernández-Fernández D, Lamas J. Antiarrhythmic calcium channel blocker verapamil inhibits trek currents in sympathetic neurons. Front Pharmacol 2022; 13:997188. [PMID: 36188584 PMCID: PMC9522527 DOI: 10.3389/fphar.2022.997188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
Background and Purpose: Verapamil, a drug widely used in certain cardiac pathologies, exert its therapeutic effect mainly through the blockade of cardiac L-type calcium channels. However, we also know that both voltage-dependent and certain potassium channels are blocked by verapamil. Because sympathetic neurons of the superior cervical ganglion (SCG) are known to express a good variety of potassium currents, and to finely tune cardiac activity, we speculated that the effect of verapamil on these SCG potassium channels could explain part of the therapeutic action of this drug. To address this question, we decided to study, the effects of verapamil on three different potassium currents observed in SCG neurons: delayed rectifier, A-type and TREK (a subfamily of K2P channels) currents. We also investigated the effect of verapamil on the electrical behavior of sympathetic SCG neurons. Experimental Approach: We employed the Patch-Clamp technique to mouse SCG neurons in culture. Key Results: We found that verapamil depolarizes of the resting membrane potential of SCG neurons. Moreover, we demonstrated that this drug also inhibits A-type potassium currents. Finally, and most importantly, we revealed that the current driven through TREK channels is also inhibited in the presence of verapamil. Conclusion and Implications: We have shown that verapamil causes a clear alteration of excitability in sympathetic nerve cells. This fact undoubtedly leads to an alteration of the sympathetic-parasympathetic balance which may affect cardiac function. Therefore, we propose that these possible peripheral alterations in the autonomic system should be taken into consideration in the prescription of this drug.
Collapse
Affiliation(s)
- S. Herrera-Pérez
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Grupo de Neurofisiología Experimental y Circuitos Neuronales, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
- *Correspondence: S. Herrera-Pérez, ; J. A. Lamas,
| | - L. Rueda-Ruzafa
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | - A. Campos-Ríos
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | | | - J.A. Lamas
- Laboratory of Neuroscience, CINBIO, University of Vigo, Vigo, Spain
- Laboratory of Neuroscience, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
- *Correspondence: S. Herrera-Pérez, ; J. A. Lamas,
| |
Collapse
|
23
|
Zou Y, Xie J, Tian W, Wu L, Xie Y, Huang S, Tang Y, Deng X, Wu H, Xie X. Integrative Analysis of KCNK Genes and Establishment of a Specific Prognostic Signature for Breast Cancer. Front Cell Dev Biol 2022; 10:839986. [PMID: 35656548 PMCID: PMC9152175 DOI: 10.3389/fcell.2022.839986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/29/2022] [Indexed: 12/19/2022] Open
Abstract
Two-pore domains potassium channel subunits, encoded by KCNK genes, play vital roles in breast cancer progression. However, the characteristics of most KCNK genes in breast cancer has yet to be clarified. In this study, we comprehensively analyzed the expression, alteration, prognosis, and biological functions of various KCNKs in breast cancer. The expression of KCNK1/4/6/9/10/13 were significantly upregulated, while KCNK2/3/5/7/17 were downregulated in breast cancer tissues compared to normal mammary tissues. Increased expression of KCNK1/3/4/9 was correlated with poor overall survival, while high expression of KCNK2/7/17 predicted better overall survival in breast cancer. Eight KCNK genes were altered in breast cancer patients with a genomic mutation rate ranged from 1.9% to 21%. KCNK1 and KCNK9 were the two most common mutations in breast cancer, occurred in 21% and 18% patients, respectively. Alteration of KCNK genes was associated with the worse clinical characteristics and higher TMB, MSI, and hypoxia score. Using machine learning method, a specific prognostic signature with seven KCNK genes was established, which manifested accuracy in predicting the prognosis of breast cancer in both training and validation cohorts. A nomogram with great predictive performance was afterwards constructed through incorporating KCNK-based risk score with clinical features. Furthermore, KCNKs were correlated with the activation of several tumor microenvironment cells, including T cells, mast cells, macrophages, and platelets. Presentation of antigen, stimulation of G protein signaling and toll-like receptor cascaded were regulated by KCNKs family. Taken together, KCNKs may regulate breast cancer progression via modulating immune response which can serve as ideal prognostic biomarkers for breast cancer patients. Our study provides novel insight for future studies evaluating their usefulness as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hao Wu
- *Correspondence: Hao Wu, ; Xinhua Xie,
| | | |
Collapse
|
24
|
Pope L, Minor DL. The Polysite Pharmacology of TREK K 2P Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:51-65. [PMID: 35138610 DOI: 10.1007/978-981-16-4254-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
K2P (KCNK) potassium channels form "background" or "leak" currents that have critical roles in cell excitability control in the brain, cardiovascular system, and somatosensory neurons. Similar to many ion channel families, studies of K2Ps have been limited by poor pharmacology. Of six K2P subfamilies, the thermo- and mechanosensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) are the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that underlie K2P function and have uncovered sites residing at every level of the channel structure with respect to the membrane where small molecules or lipids can control channel function. This polysite pharmacology within a relatively small (~70 kDa) ion channel comprises four structurally defined modulator binding sites that occur above (Keystone inhibitor site), at the level of (K2P modulator pocket), and below (Fenestration and Modulatory lipid sites) the C-type selectivity filter gate that is at the heart of K2P function. Uncovering this rich structural landscape provides the framework for understanding and developing subtype-selective modulators to probe K2P function that may provide leads for drugs for anesthesia, pain, arrhythmia, ischemia, and migraine.
Collapse
Affiliation(s)
- Lianne Pope
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, US
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, US. .,Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA. .,California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, USA. .,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, USA. .,Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
25
|
Riel EB, Jürs BC, Cordeiro S, Musinszki M, Schewe M, Baukrowitz T. The versatile regulation of K2P channels by polyanionic lipids of the phosphoinositide and fatty acid metabolism. J Gen Physiol 2022; 154:212926. [PMID: 34928298 PMCID: PMC8693234 DOI: 10.1085/jgp.202112989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/01/2021] [Indexed: 12/29/2022] Open
Abstract
Work over the past three decades has greatly advanced our understanding of the regulation of Kir K+ channels by polyanionic lipids of the phosphoinositide (e.g., PIP2) and fatty acid metabolism (e.g., oleoyl-CoA). However, comparatively little is known regarding the regulation of the K2P channel family by phosphoinositides and by long-chain fatty acid–CoA esters, such as oleoyl-CoA. We screened 12 mammalian K2P channels and report effects of polyanionic lipids on all tested channels. We observed activation of members of the TREK, TALK, and THIK subfamilies, with the strongest activation by PIP2 for TRAAK and the strongest activation by oleoyl-CoA for TALK-2. By contrast, we observed inhibition for members of the TASK and TRESK subfamilies. Our results reveal that TASK-2 channels have both activatory and inhibitory PIP2 sites with different affinities. Finally, we provided evidence that PIP2 inhibition of TASK-1 and TASK-3 channels is mediated by closure of the recently identified lower X-gate as critical mutations within the gate (i.e., L244A, R245A) prevent PIP2-induced inhibition. Our findings establish that K+ channels of the K2P family are highly sensitive to polyanionic lipids, extending our knowledge of the mechanisms of lipid regulation and implicating the metabolism of these lipids as possible effector pathways to regulate K2P channel activity.
Collapse
Affiliation(s)
- Elena B Riel
- Institute of Physiology, Kiel University, Kiel, Germany
| | - Björn C Jürs
- Institute of Physiology, Kiel University, Kiel, Germany.,Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | | | | | - Marcus Schewe
- Institute of Physiology, Kiel University, Kiel, Germany
| | | |
Collapse
|
26
|
Zheng X, Yang J, Zhu Z, Fang Y, Tian Y, Xie M, Wang W, Liu Y. The Two-Pore Domain Potassium Channel TREK-1 Promotes Blood-Brain Barrier Breakdown and Exacerbates Neuronal Death After Focal Cerebral Ischemia in Mice. Mol Neurobiol 2022; 59:2305-2327. [PMID: 35067892 DOI: 10.1007/s12035-021-02702-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022]
Abstract
Earlier studies have shown the neuroprotective role of TWIK-related K+ channel 1 (TREK-1) in global cerebral and spinal cord ischemia, while its function in focal cerebral ischemia has long been debated. This study used TREK-1-deficient mice to directly investigate the role of TREK-1 after focal cerebral ischemia. First, immunofluorescence assays in the mouse cerebral cortex indicated that TREK-1 expression was mostly abundant in astrocytes, neurons, and oligodendrocyte precursor cells but was low in myelinating oligodendrocytes, microglia, or endothelial cells. TREK-1 deficiency did not affect brain weight and morphology or the number of neurons, astrocytes, or microglia but did increase glial fibrillary acidic protein (GFAP) expression in astrocytes of the cerebral cortex. The anatomy of the major cerebral vasculature, number and structure of brain micro blood vessels, and blood-brain barrier integrity were unaltered. Next, mice underwent 60 min of focal cerebral ischemia and 72 h of reperfusion induced by the intraluminal suture method. TREK-1-deficient mice showed less neuronal death, smaller infarction size, milder blood-brain barrier (BBB) breakdown, reduced immune cell invasion, and better neurological function. Finally, the specific pharmacological inhibition of TREK-1 also decreased infarction size and improved neurological function. These results demonstrated that TREK-1 might play a detrimental rather than beneficial role in focal cerebral ischemia, and inhibition of TREK-1 would be a strategy to treat ischemic stroke in the clinic.
Collapse
Affiliation(s)
- Xiaolong Zheng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhou Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongkang Fang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yeye Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Neurological Diseases of Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
27
|
Nashed MG, Waye S, Hasan SMN, Nguyen D, Wiseman M, Zhang J, Lau H, Dinesh OC, Raymond R, Greig IR, Bambico FR, Nobrega JN. Antidepressant activity of pharmacological and genetic deactivation of the small-conductance calcium-activated potassium channel subtype-3. Psychopharmacology (Berl) 2022; 239:253-266. [PMID: 34982171 DOI: 10.1007/s00213-021-06045-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE The voltage-insensitive, small-conductance calcium-activated potassium (SK) channel is a key regulator of neuronal depolarization and is implicated in the pathophysiology of depressive disorders. OBJECTIVE We ascertained whether the SK channel is impaired in the chronic unpredictable stress (CUS) model and whether it can serve as a molecular target of antidepressant action. METHODS We assessed the depressive-like behavioral phenotype of CUS-exposed rats and performed post-mortem SK channel binding and activity-dependent zif268 mRNA analyses on their brains. To begin an assessment of SK channel subtypes involved, we examined the effects of genetic and pharmacological inhibition of the SK3 channel using conditional knockout mice and selective SK3 channel negative allosteric modulators (NAMs). RESULTS We found that [125I]apamin binding to SK channels is increased in the prefrontal cortex and decreased in the hippocampus, an effect that was associated with reciprocal levels of zif268 mRNA transcripts indicating abnormal regional cell activity in this model. We found that genetic and pharmacological manipulations significantly decreased immobility in the forced swim test without altering general locomotor activity, a hallmark of antidepressant-like activity. CONCLUSIONS Taken together, these findings link depression-related neural and behavioral pathophysiology with abnormal SK channel functioning and suggest that this can be reversed by the selective inhibition of SK3 channels.
Collapse
Affiliation(s)
- Mina G Nashed
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Shannon Waye
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Diana Nguyen
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Micaela Wiseman
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Jing Zhang
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Harry Lau
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - O Chandani Dinesh
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada
| | - Roger Raymond
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| | - Iain R Greig
- Institute of Medical Sciences, University of Aberdeen, King's College, Aberdeen, AB25 2ZD, Scotland
| | - Francis Rodriguez Bambico
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada. .,Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland & Labrador, A1B 3X9, Canada.
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Research Imaging Center, Centre for Addiction and Mental Health (CAMH), Toronto, ON, M5T 1R8, Canada
| |
Collapse
|
28
|
Kim SC, Kim A, Park JY, Hwang EM. Improved AAV vector system for cell-type-specific RNA interference. J Neurosci Methods 2021; 368:109452. [PMID: 34953938 DOI: 10.1016/j.jneumeth.2021.109452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/28/2021] [Accepted: 12/17/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND RNA interference (RNAi) is a powerful technique to effectively silence or knock down gene function in mammalian cells. For better cell-type RNAi experiments in vivo, AAV vector-based RNA interference systems need to be improved. New method: In this study, we developed an AAV vector (CREon shRNA) that expressed CRE-dependent short hairpin RNA (shRNA) and fluorescent proteins simultaneously. RESULTS We verified the Cre-dependent knockdown efficiency of the newly developed CREon shRNA vector in both HEK293T cells overexpressing TREK-1 and PC3 cells with endogenous TREK-1. Next, we packaged this TREK-1 CREon vector with AAV and injected it into the hippocampus of the brain together with a synapsin or GFAP promoter-driven CRE virus, confirming that it works well cell-selectively even in vivo. Finally, this viral vector was applied to an animal model of LPS-induced depression to determine whether behavioral changes occurred. Comparison with existing methods: With the existing pSico or pAAV-Sico-Red vectors, expression of fluorescent protein disappears when shRNA is conditionally activated by CRE recombinase, but our Creon shRNA vector showed simultaneous expression of both shRNA and fluorescent protein. Thus, it offers the advantage of allowing easy visual distinction of knocked-down cells. CONCLUSION The newly improved CREon shRNA vector can be used as a novel research tool for conditional shRNA, and may be useful for various in vivo studies such as cancer and neurobiology.
Collapse
Affiliation(s)
- Seung-Chan Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Ajung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Jae-Yong Park
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
29
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
30
|
Hashimoto K, Yamawaki Y, Yamaoka K, Yoshida T, Okada K, Tan W, Yamasaki M, Matsumoto-Makidono Y, Kubo R, Nakayama H, Kataoka T, Kanematsu T, Watanabe M, Okamoto Y, Morinobu S, Aizawa H, Yamawaki S. Spike firing attenuation of serotonin neurons in learned helplessness rats is reversed by ketamine. Brain Commun 2021; 3:fcab285. [PMID: 34939032 PMCID: PMC8688795 DOI: 10.1093/braincomms/fcab285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/04/2021] [Accepted: 10/25/2021] [Indexed: 11/14/2022] Open
Abstract
Animals suffering from uncontrollable stress sometimes show low effort to escape stress (learned helplessness). Changes in serotonin (5-hydroxytryptamine) signalling are thought to underlie this behaviour. Although the release of 5-hydroxytryptamine is triggered by the action potential firing of dorsal raphe nuclei 5-hydroxytryptamine neurons, the electrophysiological changes induced by uncontrollable stress are largely unclear. Herein, we examined electrophysiological differences among 5-hydroxytryptamine neurons in naïve rats, learned helplessness rats and rats resistant to inescapable stress (non-learned helplessness). Five-week-old male Sprague Dawley rats were exposed to inescapable foot shocks. After an avoidance test session, rats were classified as learned helplessness or non-learned helplessness. Activity-dependent 5-hydroxytryptamine release induced by the administration of high-potassium solution was slower in free-moving learned helplessness rats. Subthreshold electrophysiological properties of 5-hydroxytryptamine neurons were identical among the three rat groups, but the depolarization-induced spike firing was significantly attenuated in learned helplessness rats. To clarify the underlying mechanisms, potassium (K+) channels regulating the spike firing were initially examined using naïve rats. K+ channels sensitive to 500 μM tetraethylammonium caused rapid repolarization of the action potential and the small conductance calcium-activated K+ channels produced afterhyperpolarization. Additionally, dendrotoxin-I, a blocker of Kv1.1 (encoded by Kcna1), Kv1.2 (encoded by Kcna2) and Kv1.6 (encoded by Kcna6) voltage-dependent K+ channels, weakly enhanced the spike firing frequency during depolarizing current injections without changes in individual spike waveforms in naïve rats. We found that dendrotoxin-I significantly enhanced the spike firing of 5-hydroxytryptamine neurons in learned helplessness rats. Consequently, the difference in spike firing among the three rat groups was abolished in the presence of dendrotoxin-I. These results suggest that the upregulation of dendrotoxin-I-sensitive Kv1 channels underlies the firing attenuation of 5-hydroxytryptamine neurons in learned helplessness rats. We also found that the antidepressant ketamine facilitated the spike firing of 5-hydroxytryptamine neurons and abolished the firing difference between learned helplessness and non-learned helplessness by suppressing dendrotoxin-I-sensitive Kv1 channels. The dendrotoxin-I-sensitive Kv1 channel may be a potential target for developing drugs to control activity of 5-hydroxytryptamine neurons.
Collapse
Affiliation(s)
- Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yosuke Yamawaki
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Kenji Yamaoka
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takayuki Yoshida
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Kana Okada
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Wanqin Tan
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yoshiko Matsumoto-Makidono
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Reika Kubo
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hisako Nakayama
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Tsutomu Kataoka
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Takashi Kanematsu
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yasumasa Okamoto
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shigeru Morinobu
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shigeto Yamawaki
- Department of Psychiatry and Neurosciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| |
Collapse
|
31
|
Xie B, Hao C, Sun R. The impact of fluoxetine and pH values on relaxation of the ternary lipid monolayers. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183760. [PMID: 34499884 DOI: 10.1016/j.bbamem.2021.183760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/23/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
Fluoxetine (FLX), used in the clinic to treat depression, is a well-known cationic amphiphilic antidepressant. To get a deeper insight into the effect of FLX on Langmuir monolayers, in this study the stability and relaxation of 1,2-dioctadecanoyl-sn-glycero-3-phophocholine/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine/cholesterol (DSPC/POPC/CHOL) monolayers without and with FLX at different pH values were studied. The experiments involved surface pressure-area (π-A) measurements, mean molecular area-time (A-t) measurements, and atomic force microscope (AFM) analysis. It was found that intermolecular interactions decreased after the addition of FLX in the subphase but increased with increasing pH values. The relaxation of the ternary lipid monolayers with FLX was dominated by dissolution steps, and the dissolution rates decreased with increasing pH values. These findings can be easily confirmed by the analysis of thermodynamic parameters calculated for the investigated films. The data obtained in this study help to understand the effect of drugs on the ternary lipid monolayers from the molecular point of view.
Collapse
Affiliation(s)
- Bin Xie
- School of Physics and Electrical Engineering, Kashi University, Kashi 844009, PR China.
| | - Changchun Hao
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, PR China
| | - Runguang Sun
- School of Physics and Information Technology, Shaanxi Normal University, Xi'an 710062, PR China
| |
Collapse
|
32
|
Mini-Review: Two Brothers in Crime - The Interplay of TRESK and TREK in Human Diseases. Neurosci Lett 2021; 769:136376. [PMID: 34852287 DOI: 10.1016/j.neulet.2021.136376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 02/07/2023]
Abstract
TWIK-related spinal cord potassium (TRESK) and TWIK-related potassium (TREK) channels are both subfamilies of the two-pore domain potassium (K2P) channel group. Despite major structural, pharmacological, as well as biophysical differences, emerging data suggest that channels of these two subfamilies are functionally more closely related than previously assumed. Recent studies, for instance, indicate an assembling of TRESK and TREK subunits, leading to the formation of heterodimeric channels with different functional properties compared to homodimeric ones. Formation of tandems consisting of TRESK and TREK subunits might thus multiply the functional diversity of both TRESK and TREK activity. Based on the involvement of these channels in the pathophysiology of migraine, we here highlight the role as well as the impact of the interplay of TRESK and TREK subunits in the context of different disease settings. In this regard, we focus on their involvement in migraine and pain syndromes, as well as on their influence on (neuro-)inflammatory processes. Furthermore, we describe the potential implications for innovative therapeutic strategies that take advantage of TRESK and TREK modulation as well as obstacles encountered in the development of therapies related to the aforementioned diseases.
Collapse
|
33
|
Activity of TREK-2-like Channels in the Pyramidal Neurons of Rat Medial Prefrontal Cortex Depends on Cytoplasmic Calcium. BIOLOGY 2021; 10:biology10111119. [PMID: 34827112 PMCID: PMC8614805 DOI: 10.3390/biology10111119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
Simple Summary The pyramidal neurons of rat prefrontal cortex express potassium channels identified as a non-canonical splice variant of the TREK-2 channel. The main function of TREK channels is to regulate the resting membrane potential. We showed that cytoplasmic Ca2+ upregulates the activity of TREK-2-like channels. Previous studies have indicated that the activation of TREK-2 channels is mediated by PI(4,5)P2, a polyanionic lipid in the inner leaflet of the plasma membrane. While TREK channels are believed to not be regulated by calcium, our work shows otherwise. We propose a model in which calcium ions enable the formation of PI(4,5)P2 nanoclusters, which stabilize active conformation of the channel. Abstract TREK-2-like channels in the pyramidal neurons of rat prefrontal cortex are characterized by a wide range of spontaneous activity—from very low to very high—independent of the membrane potential and the stimuli that are known to activate TREK-2 channels, such as temperature or membrane stretching. The aim of this study was to discover what factors are involved in high levels of TREK-2-like channel activity in these cells. Our research focused on the PI(4,5)P2-dependent mechanism of channel activity. Single-channel patch clamp recordings were performed on freshly dissociated pyramidal neurons of rat prefrontal cortexes in both the cell-attached and inside-out configurations. To evaluate the role of endogenous stimulants, the activity of the channels was recorded in the presence of a PI(4,5)P2 analogue (PI(4,5)P2DiC8) and Ca2+. Our research revealed that calcium ions are an important factor affecting TREK-2-like channel activity and kinetics. The observation that calcium participates in the activation of TREK-2-like channels is a new finding. We showed that PI(4,5)P2-dependent TREK-2 activity occurs when the conditions for PI(4,5)P2/Ca2+ nanocluster formation are met. We present a possible model explaining the mechanism of calcium action.
Collapse
|
34
|
Choveau FS, Ben Soussia I, Bichet D, Franck CC, Feliciangeli S, Lesage F. Convergence of Multiple Stimuli to a Single Gate in TREK1 and TRAAK Potassium Channels. Front Pharmacol 2021; 12:755826. [PMID: 34658895 PMCID: PMC8514629 DOI: 10.3389/fphar.2021.755826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022] Open
Abstract
Inhibitory potassium channels of the TREK1/TRAAK family are integrators of multiple stimuli, including temperature, membrane stretch, polyunsaturated fatty acids and pH. How these signals affect the gating of these channels is the subject of intense research. We have previously identified a cytoplasmic domain, pCt, which plays a major role in controlling channel activity. Here, we use pharmacology to show that the effects of pCt, arachidonic acid, and extracellular pH converge to the same gate within the channel. Using a state-dependent inhibitor, fluoxetine, as well as natural and synthetic openers, we provide further evidence that the “up” and “down” conformations identified by crystallography do not correspond to open and closed states of these channels.
Collapse
Affiliation(s)
- Frank S Choveau
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Ismail Ben Soussia
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Delphine Bichet
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Chatelain C Franck
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Sylvain Feliciangeli
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Florian Lesage
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| |
Collapse
|
35
|
Conrad LJ, Proks P, Tucker SJ. Effects of ionic strength on gating and permeation of TREK-2 K2P channels. PLoS One 2021; 16:e0258275. [PMID: 34618865 PMCID: PMC8496810 DOI: 10.1371/journal.pone.0258275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/22/2021] [Indexed: 11/26/2022] Open
Abstract
In addition to the classical voltage-dependent behavior mediated by the voltage-sensing-domains (VSD) of ion channels, a growing number of voltage-dependent gating behaviors are being described in channels that lack canonical VSDs. A common thread in their mechanism of action is the contribution of the permeating ion to this voltage sensing process. The polymodal K2P K+ channel, TREK2 responds to membrane voltage through a gating process mediated by the interaction of K+ with its selectivity filter. Recently, we found that this action can be modulated by small molecule agonists (e.g. BL1249) which appear to have an electrostatic influence on K+ binding within the inner cavity and produce an increase in the single-channel conductance of TREK-2 channels. Here, we directly probed this K+-dependent gating process by recording both macroscopic and single-channel currents of TREK-2 in the presence of high concentrations of internal K+. Surprisingly we found TREK-2 is inhibited by high internal K+ concentrations and that this is mediated by the concomitant increase in ionic-strength. However, we were still able to determine that the increase in single channel conductance in the presence of BL1249 was blunted in high ionic-strength, whilst its activatory effect (on channel open probability) persisted. These effects are consistent with an electrostatic mechanism of action of negatively charged activators such as BL1249 on permeation, but also suggest that their influence on channel gating is complex.
Collapse
Affiliation(s)
- Linus J. Conrad
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, United Kingdom
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, United Kingdom
| | - Peter Proks
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| | - Stephen J. Tucker
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, United Kingdom
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Rietmeijer RA, Sorum B, Li B, Brohawn SG. Physical basis for distinct basal and mechanically gated activity of the human K + channel TRAAK. Neuron 2021; 109:2902-2913.e4. [PMID: 34390650 PMCID: PMC8448962 DOI: 10.1016/j.neuron.2021.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
TRAAK is a mechanosensitive two-pore domain K+ (K2P) channel localized to nodes of Ranvier in myelinated neurons. TRAAK deletion in mice results in mechanical and thermal allodynia, and gain-of-function mutations cause the human neurodevelopmental disorder FHEIG. TRAAK displays basal and stimulus-gated activities typical of K2Ps, but the mechanistic and structural differences between these modes are unknown. Here, we demonstrate that basal and mechanically gated openings are distinguished by their conductance, kinetics, and structure. Basal openings are low conductance, short duration, and due to a conductive channel conformation with the interior cavity exposed to the surrounding membrane. Mechanically gated openings are high conductance, long duration, and due to a channel conformation in which the interior cavity is sealed to the surrounding membrane. Our results explain how dual modes of activity are produced by a single ion channel and provide a basis for the development of state-selective pharmacology with the potential to treat disease.
Collapse
Affiliation(s)
- Robert A Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Program, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Baobin Li
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
37
|
Contribution of Neuronal and Glial Two-Pore-Domain Potassium Channels in Health and Neurological Disorders. Neural Plast 2021; 2021:8643129. [PMID: 34434230 PMCID: PMC8380499 DOI: 10.1155/2021/8643129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023] Open
Abstract
Two-pore-domain potassium (K2P) channels are widespread in the nervous system and play a critical role in maintaining membrane potential in neurons and glia. They have been implicated in many stress-relevant neurological disorders, including pain, sleep disorder, epilepsy, ischemia, and depression. K2P channels give rise to leaky K+ currents, which stabilize cellular membrane potential and regulate cellular excitability. A range of natural and chemical effectors, including temperature, pressure, pH, phospholipids, and intracellular signaling molecules, substantially modulate the activity of K2P channels. In this review, we summarize the contribution of K2P channels to neuronal excitability and to potassium homeostasis in glia. We describe recently discovered functions of K2P channels in glia, such as astrocytic passive conductance and glutamate release, microglial surveillance, and myelin generation by oligodendrocytes. We also discuss the potential role of glial K2P channels in neurological disorders. In the end, we discuss current limitations in K2P channel researches and suggest directions for future studies.
Collapse
|
38
|
Proks P, Schewe M, Conrad LJ, Rao S, Rathje K, Rödström KEJ, Carpenter EP, Baukrowitz T, Tucker SJ. Norfluoxetine inhibits TREK-2 K2P channels by multiple mechanisms including state-independent effects on the selectivity filter gate. J Gen Physiol 2021; 153:212184. [PMID: 34032848 PMCID: PMC8155809 DOI: 10.1085/jgp.202012812] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/06/2021] [Indexed: 12/25/2022] Open
Abstract
The TREK subfamily of two-pore domain K+ (K2P) channels are inhibited by fluoxetine and its metabolite, norfluoxetine (NFx). Although not the principal targets of this antidepressant, TREK channel inhibition by NFx has provided important insights into the conformational changes associated with channel gating and highlighted the role of the selectivity filter in this process. However, despite the availability of TREK-2 crystal structures with NFx bound, the precise mechanisms underlying NFx inhibition remain elusive. NFx has previously been proposed to be a state-dependent inhibitor, but its binding site suggests many possible ways in which this positively charged drug might inhibit channel activity. Here we show that NFx exerts multiple effects on single-channel behavior that influence both the open and closed states of the channel and that the channel can become highly activated by 2-APB while remaining in the down conformation. We also show that the inhibitory effects of NFx are unrelated to its positive charge but can be influenced by agonists which alter filter stability, such as ML335, as well as by an intrinsic voltage-dependent gating process within the filter. NFx therefore not only inhibits channel activity by altering the equilibrium between up and down conformations but also can directly influence filter gating. These results provide further insight into the complex allosteric mechanisms that modulate filter gating in TREK K2P channels and highlight the different ways in which filter gating can be regulated to permit polymodal regulation.
Collapse
Affiliation(s)
- Peter Proks
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK
| | - Marcus Schewe
- Department of Physiology, University of Kiel, Kiel, Germany
| | - Linus J Conrad
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK
| | - Shanlin Rao
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Kristin Rathje
- Department of Physiology, University of Kiel, Kiel, Germany
| | | | - Elisabeth P Carpenter
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK.,Centre for Medicines Discovery, University of Oxford, UK
| | | | - Stephen J Tucker
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, UK
| |
Collapse
|
39
|
Li XL, Tang CY, Wang S, Zhao M, Wang XF, Li TF, Qi XL, Luan GM, Guan YG. Regulation of TWIK-related K + channel 1 in the anterior hippocampus of patients with temporal lobe epilepsy with comorbid depression. Epilepsy Behav 2021; 121:108045. [PMID: 34116339 DOI: 10.1016/j.yebeh.2021.108045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/11/2021] [Accepted: 04/30/2021] [Indexed: 02/08/2023]
Abstract
Epilepsy with comorbid depression has recently attracted increasing attention. Temporal lobe epilepsy (TLE) may represent an increased risk of developing depression, especially if the seizures do not generalize. The two-pore domain potassium channel-TWIK-related K+ channel (TREK-1) plays important roles in both epilepsy and depression. However, the changes in its expression in patients with epilepsy with comorbid depression remain unclear. In the present study, we analyzed depressive symptoms using neuropsychiatric scales in forty-two patients with drug-resistant TLE, who also underwent EEG in waking and sleeping states, as well as 3.0 T brain MRI. We tested for TREK-1 positive neurons and microglial cells in the anterior hippocampi of patients with drug-resistant TLE with and without comorbid depression (n=5/group). Approximately 31% of patients with TLE had comorbid depression (13/42). Meanwhile, the patients who had hippocampal sclerosis had much higher scores on the depression rating scale. The results indicated the contribution of hippocampal sclerosis to the development of depression. Immunostaining of TREK-1 channels was observed in neurons and glia in the anterior hippocampus. Increased immunoreactivity of TREK-1 neurons was observed in the hippocampi of patients with TLE with comorbid depression compared with nondepressed patients with TLE. TREK-1 was expressed in almost all microglia. Curiously, more activated TREK-1-positive microglia were observed in patients with TLE with depression than in those without depression. The results suggested that a change in TREK-1 immunoreactivity was involved, at least partly, in the development of depression as a comorbidity of TLE. Imbalance of the TREK-1 channel may be a potential target for the treatment of patients with epilepsy with comorbid depression.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Neurology, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China; Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chong-Yang Tang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Shu Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Meng Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Xiong-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tian-Fu Li
- Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Epilepsy, Beijing, China; Center of Epilepsy, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Xue-Ling Qi
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Guo-Ming Luan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Epilepsy, Beijing, China; Center of Epilepsy, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Yu-Guang Guan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Epilepsy, Beijing, China; Center of Epilepsy, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
40
|
Cunningham KP, Clapp LH, Mathie A, Veale EL. The Prostacyclin Analogue, Treprostinil, Used in the Treatment of Pulmonary Arterial Hypertension, is a Potent Antagonist of TREK-1 and TREK-2 Potassium Channels. Front Pharmacol 2021; 12:705421. [PMID: 34267666 PMCID: PMC8276018 DOI: 10.3389/fphar.2021.705421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an aggressive vascular remodeling disease that carries a high morbidity and mortality rate. Treprostinil (Remodulin) is a stable prostacyclin analogue with potent vasodilatory and anti-proliferative activity, approved by the FDA and WHO as a treatment for PAH. A limitation of this therapy is the severe subcutaneous site pain and other forms of pain experienced by some patients, which can lead to significant non-compliance. TWIK-related potassium channels (TREK-1 and TREK-2) are highly expressed in sensory neurons, where they play a role in regulating sensory neuron excitability. Downregulation, inhibition or mutation of these channels leads to enhanced pain sensitivity. Using whole-cell patch-clamp electrophysiological recordings, we show, for the first time, that treprostinil is a potent antagonist of human TREK-1 and TREK-2 channels but not of TASK-1 channels. An increase in TASK-1 channel current was observed with prolonged incubation, consistent with its therapeutic role in PAH. To investigate treprostinil-induced inhibition of TREK, site-directed mutagenesis of a number of amino acids, identified as important for the action of other regulatory compounds, was carried out. We found that a gain of function mutation of TREK-1 (Y284A) attenuated treprostinil inhibition, while a selective activator of TREK channels, BL-1249, overcame the inhibitory effect of treprostinil. Our data suggests that subcutaneous site pain experienced during treprostinil therapy may result from inhibition of TREK channels near the injection site and that pre-activation of these channels prior to treatment has the potential to alleviate this nociceptive activity.
Collapse
Affiliation(s)
- Kevin P Cunningham
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom.,Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Lucie H Clapp
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom.,School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, United Kingdom
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
41
|
Singh S, Agarwal P, Ravichandiran V. Two-Pore Domain Potassium Channel in Neurological Disorders. J Membr Biol 2021; 254:367-380. [PMID: 34169340 DOI: 10.1007/s00232-021-00189-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/26/2021] [Indexed: 01/10/2023]
Abstract
K2P channel is the leaky potassium channel that is critical to keep up the negative resting membrane potential for legitimate electrical conductivity of the excitable tissues. Recently, many substances and medication elements are discovered that could either straightforwardly or in a roundabout way influence the 15 distinctive K+ ion channels including TWIK, TREK, TASK, TALK, THIK, and TRESK. Opening and shutting of these channels or any adjustment in their conduct is thought to alter the pathophysiological condition of CNS. There is no document available till now to explain in detail about the molecular mechanism of agents acting on K2P channel. Accordingly, in this review we cover the current research and mechanism of action of these channels, we have also tried to mention the detailed effect of drugs and how the channel behavior changes by focusing on recent advances regarding activation and modulation of ion channels.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Industrial Area, Hajipur, District, Vaishali, 844102, Bihar, India.
| | - Punita Agarwal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Industrial Area, Hajipur, District, Vaishali, 844102, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Export Promotions Industrial Park (EPIP), Industrial Area, Hajipur, District, Vaishali, 844102, Bihar, India
| |
Collapse
|
42
|
Walsh Y, Leach M, Veale EL, Mathie A. Block of TREK and TRESK K2P channels by lamotrigine and two derivatives sipatrigine and CEN-092. Biochem Biophys Rep 2021; 26:101021. [PMID: 34041373 PMCID: PMC8144350 DOI: 10.1016/j.bbrep.2021.101021] [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: 03/26/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 11/30/2022] Open
Abstract
TREK and TRESK K2P channels are widely expressed in the nervous system, particularly in sensory neurons, where they regulate neuronal excitability. In this study, using whole-cell patch-clamp electrophysiology, we characterise the inhibitory effect of the anticonvulsant lamotrigine and two derivatives, sipatrigine and 3,5-diamino-6-(3,5-bistrifluoromethylphenyl)-1,2,4-triazine (CEN-092) on these channels. Sipatrigine was found to be a more effective inhibitor than lamotrigine of TREK-1, TREK-2 and TRESK channels. Sipatrigine was slightly more potent on TREK-1 channels (EC50 = 16 μM) than TRESK (EC50 = 34 μM) whereas lamotrigine was equally effective on TREK-1 and TRESK. Sipatrigine was less effective on a short isoform of TREK-2, suggesting the N terminus of the channel is important for both inhibition and subsequent over-recovery. Inhibition of TREK-1 and TREK-2 channels by sipatrigine was reduced by mutation of a leucine residue associated with the norfluoxetine binding site on these channels (L289A and L320A on TREK-1 and TREK-2, respectively) but these did not affect inhibition by lamotrigine. Inhibition of TRESK by sipatrigine and lamotrigine was attenuated by mutation of bulky phenylalanine residues (F145A and F352A) in the inner pore helix. However, phosphorylation mutations did not alter the effect of sipatrigine. CEN-092 was a more effective inhibitor of TRESK channels than TREK-1 channels. It is concluded that lamotrigine, sipatrigine and CEN-092 are all inhibitors of TREK and TRESK channels but do not greatly discriminate between them. The actions of these compounds may contribute to their current and potential use in the treatment of pain and depression. Lamotrigine blocks TREK and TRESK potassium channels at clinical concentrations. Sipatrigine is more effective than lamotrigine at blocking TREK and TRESK channels. Mutation of norfluoxetine binding site on TREK channels attenuates sipatrigine block. Residues in the inner pore region of TRESK channels regulate sipatrigine block. The novel lamotrigine derivative, CEN-092, blocks TRESK channels.
Collapse
Affiliation(s)
- Yvonne Walsh
- Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK
- University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK
| | - Michael Leach
- University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK
| | - Emma L. Veale
- Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK
- School of Engineering, Arts, Science and Technology, University of Suffolk, Waterfront Building, Neptune Quay, Ipswich, IP4 1QJ, UK
- Corresponding author.Medway School of Pharmacy, University of Kent and University of Greenwich, Central Avenue, Chatham Maritime, ME4 4TB, UK.
| |
Collapse
|
43
|
Natale AM, Deal PE, Minor DL. Structural Insights into the Mechanisms and Pharmacology of K 2P Potassium Channels. J Mol Biol 2021; 433:166995. [PMID: 33887333 PMCID: PMC8436263 DOI: 10.1016/j.jmb.2021.166995] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/10/2023]
Abstract
Leak currents, defined as voltage and time independent flows of ions across cell membranes, are central to cellular electrical excitability control. The K2P (KCNK) potassium channel class comprises an ion channel family that produces potassium leak currents that oppose excitation and stabilize the resting membrane potential in cells in the brain, cardiovascular system, immune system, and sensory organs. Due to their widespread tissue distribution, K2Ps contribute to many physiological and pathophysiological processes including anesthesia, pain, arrythmias, ischemia, hypertension, migraine, intraocular pressure regulation, and lung injury responses. Structural studies of six homomeric K2Ps have established the basic architecture of this channel family, revealed key moving parts involved in K2P function, uncovered the importance of asymmetric pinching and dilation motions in the K2P selectivity filter (SF) C-type gate, and defined two K2P structural classes based on the absence or presence of an intracellular gate. Further, a series of structures characterizing K2P:modulator interactions have revealed a striking polysite pharmacology housed within a relatively modestly sized (~70 kDa) channel. Binding sites for small molecules or lipids that control channel function are found at every layer of the channel structure, starting from its extracellular side through the portion that interacts with the membrane bilayer inner leaflet. This framework provides the basis for understanding how gating cues sensed by different channel parts control function and how small molecules and lipids modulate K2P activity. Such knowledge should catalyze development of new K2P modulators to probe function and treat a wide range of disorders.
Collapse
Affiliation(s)
- Andrew M Natale
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Parker E Deal
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience University of California, San Francisco, CA 94158, USA; Molecular Biophysics and Integrated Bio-imaging Division Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
44
|
Huang L, Xu G, Jiang R, Luo Y, Zuo Y, Liu J. Development of Non-opioid Analgesics Targeting Two-pore Domain Potassium Channels. Curr Neuropharmacol 2021; 20:16-26. [PMID: 33827408 PMCID: PMC9199554 DOI: 10.2174/1570159x19666210407152528] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Two-pore domain potassium (K2P) channels are a diverse family of potassium channels. K2P channels generate background leak potassium currents to regulate cellular excitability and are thereby involved in a wide range of neurological disorders. K2P channels are modulated by a variety of physicochemical factors such as mechanical stretch, temperature, and pH. In the the peripheral nervous system (PNS), K2P channels are widely expressed in nociceptive neurons and play a critical roles in pain perception. In this review, we summarize the recent advances in the pharmacological properties of K2P channels, with a focus on the exogenous small-molecule activators targeting K2P channels. We emphasize the subtype-selectivity, cellular and in vivo pharmacological properties of all the reported small-molecule activators. The key underlying analgesic mechanisms mediated by K2P are also summarized based on the data in the literature from studies using small-molecule activators and genetic knock-out animals. We discuss advantages and limitations of the translational perspectives of K2P in pain medicine and provide outstanding questions for future studies in the end.
Collapse
Affiliation(s)
- Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Guangyin Xu
- Department of Physiology and Neurobiology, Institute of Neuroscience, Medical College of Soochow University, Suzhou, 215123, Jiangsu. China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yuncheng Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yunxia Zuo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| |
Collapse
|
45
|
Lee H, Lolicato M, Arrigoni C, Minor DL. Production of K 2P2.1 (TREK-1) for structural studies. Methods Enzymol 2021; 653:151-188. [PMID: 34099170 DOI: 10.1016/bs.mie.2021.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
K2P (KCNK) potassium channels form 'background' or 'leak' currents that are important for controlling cell excitability in the brain, cardiovascular system, and somatosensory neurons. K2P2.1 (TREK-1) is one of the founding members of this family and one of the first well-characterized polymodal ion channels capable of responding to a variety of physical and chemical gating cues. Of the six K2P subfamilies, the thermo-and mechano-sensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) is the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that provide a framework for understanding how gating cues sensed by different channel elements converge on the K2P selectivity filter C-type gate. TREK family structural studies have also revealed numerous sites where small molecules or lipids bind and affect channel function. This rich structural landscape provides the framework for probing K2P function and for the development of new K2P-directed agents. Such molecules may be useful for affecting processes where TREK channels are important such as anesthesia, pain, arrythmia, ischemia, migraine, intraocular pressure, and lung injury. Production of high quality protein samples is key to addressing new questions about K2P function and pharmacology. Here, we present methods for producing pure K2P2.1 (TREK-1) suitable for advancing towards these goals through structural and biochemical studies.
Collapse
Affiliation(s)
- Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Cristina Arrigoni
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, United States; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, United States; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, United States; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
46
|
Giannoni-Guzmán MA, Kamitakahara A, Magalong V, Levitt P, McMahon DG. Circadian photoperiod alters TREK-1 channel function and expression in dorsal raphe serotonergic neurons via melatonin receptor 1 signaling. J Pineal Res 2021; 70:e12705. [PMID: 33210730 PMCID: PMC8496951 DOI: 10.1111/jpi.12705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
Seasonal day length has been linked to the prevalence of mood disorders, and however, the mechanisms underlying this relationship remain unknown. Previous work in our laboratory has shown that developmental exposure to seasonal photoperiods has enduring effects on the activity of mouse dorsal raphe serotonergic neurons, their intrinsic electrical properties, as well as on depression and anxiety-related behaviors. Here we focus on the possible ionic mechanisms that underlie the observed programming of the electrophysiological properties of serotonin neurons, focusing on the twin-pore K + channels TREK-1 and TASK-1 that set resting membrane potential and regulate excitability. Pharmacological inhibition of TREK-1 significantly increased spike frequency in Short and Equinox photoperiods, but did not further elevate the firing rate in slices from Long photoperiod mice, suggesting that TREK-1 function is reduced in Long photoperiods. In contrast, inhibition of TASK-1 resulted in increases in firing rates across all photoperiods, suggesting that it contributes to setting excitability, but is not regulated by photoperiod. We then quantified Kcnk2 mRNA levels specifically in dorsal raphe 5-HT neurons using triple-label RNAscope. We found that Long photoperiod significantly reduced levels of Kcnk2 in serotonin neurons co-expressing Tph2, and Pet-1. Photoperiodic effects on the function and expression of TREK-1 were blocked in melatonin 1 receptor knockout (MT-1KO) mice, consistent with previous findings that MT-1 signaling is necessary for photoperiodic programming of dorsal raphe 5-HT neurons. Taken together these results indicate that photoperiodic regulation of TREK-1 expression and function plays a key role in photoperiodic programming the excitability of dorsal raphe 5-HT neurons.
Collapse
Affiliation(s)
| | - Anna Kamitakahara
- Department of Pediatrics and Program in Developmental Neuroscience and Neurogenetics, Keck School of Medicine, The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Valerie Magalong
- Department of Pediatrics and Program in Developmental Neuroscience and Neurogenetics, Keck School of Medicine, The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Department of Pediatrics and Program in Developmental Neuroscience and Neurogenetics, Keck School of Medicine, The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Douglas G. McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
47
|
Bae Y, Choi JH, Ryoo K, Kim A, Kwon O, Jung HG, Hwang EM, Park JY. Spadin Modulates Astrocytic Passive Conductance via Inhibition of TWIK-1/TREK-1 Heterodimeric Channels. Int J Mol Sci 2020; 21:ijms21249639. [PMID: 33348878 PMCID: PMC7765948 DOI: 10.3390/ijms21249639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Astrocytes, the most abundant cell type in the brain, are non-excitable cells and play critical roles in brain function. Mature astrocytes typically exhibit a linear current-voltage relationship termed passive conductance, which is believed to enable astrocytes to maintain potassium homeostasis in the brain. We previously demonstrated that TWIK-1/TREK-1 heterodimeric channels mainly contribute to astrocytic passive conductance. However, the molecular identity of astrocytic passive conductance is still controversial and needs to be elucidated. Here, we report that spadin, an inhibitor of TREK-1, can dramatically reduce astrocytic passive conductance in brain slices. A series of gene silencing experiments demonstrated that spadin-sensitive currents are mediated by TWIK-1/TREK-1 heterodimeric channels in cultured astrocytes and hippocampal astrocytes from brain slices. Our study clearly showed that TWIK-1/TREK-1-heterodimeric channels can act as the main molecular machinery of astrocytic passive conductance, and suggested that spadin can be used as a specific inhibitor to control astrocytic passive conductance.
Collapse
Affiliation(s)
- Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Jae Hyouk Choi
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Kanghyun Ryoo
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Ajung Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Osung Kwon
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Hyun-Gug Jung
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
- Correspondence: (E.M.H.); (J.-Y.P.)
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
- Correspondence: (E.M.H.); (J.-Y.P.)
| |
Collapse
|
48
|
Diz-Chaves Y, Herrera-Pérez S, González-Matías LC, Lamas JA, Mallo F. Glucagon-Like Peptide-1 (GLP-1) in the Integration of Neural and Endocrine Responses to Stress. Nutrients 2020; 12:nu12113304. [PMID: 33126672 PMCID: PMC7692797 DOI: 10.3390/nu12113304] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Glucagon like-peptide 1 (GLP-1) within the brain is produced by a population of preproglucagon neurons located in the caudal nucleus of the solitary tract. These neurons project to the hypothalamus and another forebrain, hindbrain, and mesolimbic brain areas control the autonomic function, feeding, and the motivation to feed or regulate the stress response and the hypothalamic-pituitary-adrenal axis. GLP-1 receptor (GLP-1R) controls both food intake and feeding behavior (hunger-driven feeding, the hedonic value of food, and food motivation). The activation of GLP-1 receptors involves second messenger pathways and ionic events in the autonomic nervous system, which are very relevant to explain the essential central actions of GLP-1 as neuromodulator coordinating food intake in response to a physiological and stress-related stimulus to maintain homeostasis. Alterations in GLP-1 signaling associated with obesity or chronic stress induce the dysregulation of eating behavior. This review summarized the experimental shreds of evidence from studies using GLP-1R agonists to describe the neural and endocrine integration of stress responses and feeding behavior.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| | - Salvador Herrera-Pérez
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | | | - José Antonio Lamas
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | - Federico Mallo
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| |
Collapse
|
49
|
Busserolles J, Ben Soussia I, Pouchol L, Marie N, Meleine M, Devilliers M, Judon C, Schopp J, Clémenceau L, Poupon L, Chapuy E, Richard S, Noble F, Lesage F, Ducki S, Eschalier A, Lolignier S. TREK1 channel activation as a new analgesic strategy devoid of opioid adverse effects. Br J Pharmacol 2020; 177:4782-4795. [PMID: 32851651 DOI: 10.1111/bph.15243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/06/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioids are effective painkillers. However, their risk-benefit ratio is dampened by numerous adverse effects and opioid misuse has led to a public health crisis. Safer alternatives are required, but isolating the antinociceptive effect of opioids from their adverse effects is a pharmacological challenge because activation of the μ opioid receptor triggers both the antinociceptive and adverse effects of opioids. EXPERIMENTAL APPROACH The TREK1 potassium channel is activated downstream of μ receptor and involved in the antinociceptive activity of morphine but not in its adverse effects. Bypassing the μ opioid receptor to directly activate TREK1 could therefore be a safer analgesic strategy. KEY RESULTS We developed a selective TREK1 activator, RNE28, with antinociceptive activity in naive rodents and in models of inflammatory and neuropathic pain. This activity was lost in TREK1 knockout mice or wild-type mice treated with the TREK1 blocker spadin, showing that TREK1 is required for the antinociceptive activity of RNE28. RNE28 did not induce respiratory depression, constipation, rewarding effects, or sedation at the analgesic doses tested. CONCLUSION AND IMPLICATIONS This proof-of-concept study shows that TREK1 activators could constitute a novel class of painkillers, inspired by the mechanism of action of opioids but devoid of their adverse effects.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Ismail Ben Soussia
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Université Côte d'Azur, INSERM, Valbonne, France
| | - Laetitia Pouchol
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Nicolas Marie
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Mathieu Meleine
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Maïly Devilliers
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Céline Judon
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Julien Schopp
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Loïc Clémenceau
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Laura Poupon
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Eric Chapuy
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Serge Richard
- Centre de Recherches Biologiques, CERB, Baugy, France
| | - Florence Noble
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Florian Lesage
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Université Côte d'Azur, INSERM, Valbonne, France
| | - Sylvie Ducki
- ICCF, SIGMA Clermont, Université Clermont Auvergne, CNRS, Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| |
Collapse
|
50
|
Francis-Oliveira J, Shieh IC, Vilar Higa GS, Barbosa MA, De Pasquale R. Maternal separation induces changes in TREK-1 and 5HT 1A expression in brain areas involved in the stress response in a sex-dependent way. Behav Brain Res 2020; 396:112909. [PMID: 32949645 DOI: 10.1016/j.bbr.2020.112909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/09/2020] [Accepted: 09/13/2020] [Indexed: 12/25/2022]
Abstract
Depression is a prevalent disease in modern society, and has been linked to stressful events at early ages. Women are more susceptible to depression, and the neural basis for this are still under investigation. Serotonin is known to be involved in depression, and a decrease in 5HT1A expression is observed on temporal and cortical areas in both men and women with depression. As knockout animals for TREK-1 are resilient to depression, this channel has emerged as a new potential pharmacological target for depression treatment. In this study, maternal separation (MS) was used to emulate early-life stress, and evaluate behaviour, as well as TREK-1 and 5HT1A expression in the brain using immunohistochemistry. In juvenile females, 5HT1A reduction coupled to increased TREK-1 in the dentate gyrus (DG) was associated with behavioural despair, as well as increased TREK-1 expression in basolateral amygdala (BLA) and prelimbic cortex (PL). In juvenile males, MS induced an increase in 5HT1A in the BLA, and in TREK-1 in the PL, while no behavioural despair was observed. Anhedonia and anxiety-like behaviour were not induced by MS. We conclude stress-induced increase in TREK-1 in PL and GD is associated to depression, while 5HT1A changes coupled to TREK-1 changes may be necessary to induce depression, with females being more vulnerable to MS effects than males. Thus, TREK-1 and 5HT1A may be potential pharmacological targets for antidepressants development.
Collapse
Affiliation(s)
- J Francis-Oliveira
- Dept of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, Ave Lineu Prestes 1524, 05508-000, São Paulo, SP, Brazil.
| | - I C Shieh
- Dept of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, Ave Lineu Prestes 1524, 05508-000, São Paulo, SP, Brazil
| | - G S Vilar Higa
- Neurogenetics Laboratory, Mathematics Computation Cognition Center, Rua Arcturus 03, 09606-070, São Bernardo do Campo, SP, Brazil
| | - M A Barbosa
- Dept of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, Ave Lineu Prestes 1524, 05508-000, São Paulo, SP, Brazil
| | - R De Pasquale
- Dept of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, Ave Lineu Prestes 1524, 05508-000, São Paulo, SP, Brazil
| |
Collapse
|