1
|
Yashiro K, Iwaki Y, Urata H, Kokubo M, Mori T, Sekioka Y, Isami K, Kato J, Wieting J, McGowan KM, Bridges TM, Boutaud O, Engers DW, Denton JS, Kurata H, Lindsley CW. Discovery of ONO-2920632 (VU6011887): A Highly Selective and CNS Penetrant TREK-2 (TWIK-Related K+ Channel 2) Preferring Activator In Vivo Tool Compound. ACS Chem Neurosci 2025; 16:960-967. [PMID: 39981749 PMCID: PMC11887051 DOI: 10.1021/acschemneuro.5c00032] [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: 01/14/2025] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Herein we describe our initial work on the K2P family of potassium ion channels with the chemical optimization and characterization of a novel series of TWIK-Related K+ Channel (TREK)-1/2 dual activators and TREK-2 preferring activators derived from a high-throughput screening hit. The exercise provided TREK activators with good CNS penetration and others with low CNS exposure to enable exploration of both central and peripheral TREK activation. From this, ONO-2920632 (VU6011887 = 19b) emerged as a reasonably potent (human Tl+; TREK-1 EC50 = 2.8 μM (95% Emax), TREK-2 EC50 = 0.30 μM (184% Emax)), first-generation CNS penetrant (rat Kp = 0.37) in vivo tool compound with selectivity versus the other K2P channels (>91-fold selective vs TASK1, TASK2, TASK3, TRAAK, TWIK2, and 31-fold selective vs TRESK) and no significant activity in a large ancillary pharmacology panel. ONO-2920632 (VU6011887) displayed robust, dose dependent efficacy when dosed orally in a mouse pain model (acetic acid writhing assay), where it was equipotent at 3 mg/kg to the assay standard indomethacin at 10 mg/kg. The therapeutic potential of TREK channel activation has long been hampered by a lack of selective, small molecule tools, and this work provides a variety of in vivo tool compounds for the community.
Collapse
Affiliation(s)
- Kentaro Yashiro
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Yuzo Iwaki
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Hirohito Urata
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Masaya Kokubo
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Takahiro Mori
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Yoko Sekioka
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Koichi Isami
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Junya Kato
- Pharmacokinetic
Research, Ono Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Joshua Wieting
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Kevin M. McGowan
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Olivier Boutaud
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Darren W. Engers
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jerod S. Denton
- Department
of Anesthesiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
| | - Haruto Kurata
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Craig W. Lindsley
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Chemistry, Vanderbilt University, Nashville Tennessee 37232, United States
- Department
of Biochemistry, Vanderbilt University, Nashville Tennessee 37232, United States
| |
Collapse
|
2
|
Roy-Chowdhury S, Jang S, Abderemane-Ali F, Naughton F, Grabe M, Minor DL. Structure of the human K 2P13.1 channel reveals a hydrophilic pore restriction and lipid cofactor site. Nat Struct Mol Biol 2025:10.1038/s41594-024-01476-3. [PMID: 40011746 DOI: 10.1038/s41594-024-01476-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/17/2024] [Indexed: 02/28/2025]
Abstract
Polyunsaturated fatty acid (PUFA) lipids modulate the neuronal and microglial leak potassium channel K2P13.1 (THIK1) and other voltage-gated ion channel (VGIC) superfamily members through poorly understood mechanisms. Here we present cryo-electron microscopy structures of human THIK1 and mutants, revealing a unique two-chamber aqueous inner cavity obstructed by a hydrophilic barrier important for gating, the flow restrictor, and a P1-M4 intersubunit interface lipid at a site, the PUFA site, corresponding to the K2P small-molecule modulator pocket. This overlap, together with functional studies, indicates that PUFA site lipids are THIK1 cofactors. Comparison with a PUFA-responsive VGIC, Kv7.1, reveals a shared modulatory role for the pore domain intersubunit interface, providing a framework for understanding PUFA action on the VGIC superfamily. Our findings reveal the distinct THIK1 architecture, highlight the importance of the P1-M4 interface for K2P control by natural and synthetic ligands and should aid in the development of THIK subfamily modulators for neuroinflammation and autism.
Collapse
Affiliation(s)
| | - Seil Jang
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Fiona Naughton
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
| | - Michael Grabe
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, UCSF Medical Center, San Francisco, CA, USA.
- Departments of Biochemistry and Biophysics and Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
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; 19:e202400409. [PMID: 39145995 PMCID: PMC11617644 DOI: 10.1002/cmdc.202400409] [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: 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 BiochemistryKangwon National UniversityCollege of Natural SciencesChuncheon24341Republic of Korea
| | - Nhung Thi Hong Van
- Department of PhysiologyDongguk UniversityCollege of MedicineGyeongju38066Republic of Korea
| | - Joo Hyun Nam
- Department of PhysiologyDongguk UniversityCollege of MedicineGyeongju38066Republic of Korea
| | - Wook Lee
- Department of BiochemistryKangwon National UniversityCollege of Natural SciencesChuncheon24341Republic of Korea
| |
Collapse
|
4
|
Torun IE, Kilinc YB, Kilinc E, Töre F. TRESK channel activation ameliorates migraine-like pain via modulation of CGRP release from the trigeminovascular system and meningeal mast cells in experimental migraine models. Life Sci 2024; 357:123091. [PMID: 39362587 DOI: 10.1016/j.lfs.2024.123091] [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: 07/23/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
AIMS Accumulating evidence indicates the involvement of TRESK potassium channels in migraine, however, effects of TRESK activation on migraine-related mechanisms remain unclear. We explored effects of TRESK channel modulation on migraine-related behavioral and molecular markers in in-vivo and ex-vivo rat models of migraine. MAIN METHODS The selective TRESK activator cloxyquin at different doses, the TRESK inhibitor A2764, and the migraine drug sumatriptan were tested alone or in different combinations in nitroglycerin (NTG)-induced in-vivo model, and in ex-vivo meningeal, trigeminal ganglion and brainstem preparations in which CGRP release was induced by capsaicin. Mechanical allodynia, CGRP and c-fos levels in trigeminovascular structures and meningeal mast cells were evaluated. KEY FINDINGS Cloxyquin attenuated NTG-induced mechanical allodynia, brainstem c-fos and CGRP levels, trigeminal ganglion CGRP levels and meningeal mast cell degranulation and number, in-vivo. It also diminished capsaicin-induced CGRP release from ex-vivo meningeal, trigeminal ganglion and brainstem preparations. Specific TRESK inhibitor A2764 abolished all effects of cloxyquin in in-vivo and ex-vivo. Combining cloxyquin and sumatriptan exerted a synergistic effect ex-vivo, but not in-vivo. SIGNIFICANCE Our findings provide the experimental evidence for the anti-migraine effect of TRESK activation in migraine-like conditions. The modulation of TRESK channels may therefore be an attractive alternative strategy to relieve migraine pain.
Collapse
Affiliation(s)
- Ibrahim Ethem Torun
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Türkiye
| | - Yasemin Baranoglu Kilinc
- Department of Pediatrics, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Türkiye
| | - Erkan Kilinc
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Türkiye.
| | - Fatma Töre
- Department of Physiology, Faculty of Medicine, Istanbul Atlas University, Istanbul, Türkiye
| |
Collapse
|
5
|
Zhang Y, Li J, Pan J, Deng S. Research progress of two-pore potassium channel in myocardial ischemia-reperfusion injury. Front Physiol 2024; 15:1473501. [PMID: 39534859 PMCID: PMC11554511 DOI: 10.3389/fphys.2024.1473501] [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: 07/31/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a secondary injury caused by restoring blood flow after acute myocardial infarction, which may lead to serious arrhythmia and heart damage. In recent years, the role of potassium channels in MIRI has attracted much attention, especially the members of the two-pore domain potassium (K2P) channel family. K2P channel has unique structure and function, and the formation of its heterodimer increases its functional diversity. This paper reviews the structural characteristics, types, expression and physiological functions of K2P channel in the heart. In particular, we pay attention to whether members of the subfamily such as TWIK, TREK, TASK, TALK, THIK and TRESK participate in MIRI and their related mechanisms. Future research will help to reveal the molecular mechanism of K2P channel in MIRI and provide new strategies for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Shengli Deng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
6
|
Wu Z, Zhu Y, Xu L, Lai W, Chen X, Long H. Development of a novel three-dimensional injection guide for trigeminal ganglia. J Neurosci Methods 2024; 409:110197. [PMID: 38878976 DOI: 10.1016/j.jneumeth.2024.110197] [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: 12/20/2023] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Trigeminal ganglion (TG) plays an important role in the process of orthodontic pain. It's necessary to design an accurate, precise and minimally invasive trigeminal ganglion injection guide plate to study TG. METHODS Micro-CT was used to obtain the Dicom format data, and three-dimensional (3D) software (mimics and magics23.03) was used to reconstruct 3D head models. Design and modifications of the TG injection guide plate were performed in Magic 23.03 software, and the guide plate was produced by a 3D stereolithography printer. X-ray, micro-CT, Evans blue, and virus transduction were used to demonstrate the accuracy of the guide-assisted injection. Pain levels were evaluated after using the injection guide by a bite force test and Von Frey test. RESULTS X-ray and micro-CT tests confirmed that the injection needle reached the bilateral trigeminal ganglia fossa. The Evans blue test and virus transduction proved that the injected drug could be accurately injected into the bilateral trigeminal ganglion and the lentivirus could be successfully transfected. The percentage of accurate injection was 10/10 (bilateral trigeminal ganglia). Orofacial pain induced by the trigeminal ganglion injection was mild and returned to baseline within seven days. CONCLUSION The injection guide described in this study is viable and reliable for the delivery of drugs and virus transduction into the trigeminal ganglia.
Collapse
Affiliation(s)
- Zhouqiang Wu
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, PR China; Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province & Key Laboratory of Oral Biomedical Research of Zhejiang Province & Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yafen Zhu
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, PR China; Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province & Key Laboratory of Oral Biomedical Research of Zhejiang Province & Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Lehan Xu
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province & Key Laboratory of Oral Biomedical Research of Zhejiang Province & Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Wenli Lai
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Xuepeng Chen
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine & Clinical Research Center for Oral Diseases of Zhejiang Province & Key Laboratory of Oral Biomedical Research of Zhejiang Province & Cancer Center of Zhejiang University, Hangzhou 310006, China.
| | - Hu Long
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, PR China.
| |
Collapse
|
7
|
Pergel E, Tóth DJ, Baukál D, Veres I, Czirják G. The Ubiquitin Ligase Adaptor NDFIP1 Interacts with TRESK and Negatively Regulates the Background K + Current. Int J Mol Sci 2024; 25:8879. [PMID: 39201565 PMCID: PMC11355008 DOI: 10.3390/ijms25168879] [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: 07/24/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
The TRESK (K2P18.1, KCNK18) background potassium channel is expressed in primary sensory neurons and has been reported to contribute to the regulation of pain sensations. In the present study, we examined the interaction of TRESK with NDFIP1 (Nedd4 family-interacting protein 1) in the Xenopus oocyte expression system by two-electrode voltage clamp and biochemical methods. We showed that the coexpression of NDFIP1 abolished the TRESK current under the condition where the other K+ channels were not affected. Mutations in the three PPxY motifs of NDFIP1, which are responsible for the interaction with the Nedd4 ubiquitin ligase, prevented a reduction in the TRESK current. Furthermore, the overexpression of a dominant-negative Nedd4 construct in the oocytes coexpressing TRESK with NDFIP1 partially reversed the down-modulating effect of the adaptor protein on the K+ current. The biochemical data were also consistent with the functional results. An interaction between epitope-tagged versions of TRESK and NDFIP1 was verified by co-immunoprecipitation experiments. The coexpression of NDFIP1 with TRESK induced the ubiquitination of the channel protein. Altogether, the results suggest that TRESK is directly controlled by and highly sensitive to the activation of the NDFIP1-Nedd4 system. The NDFIP1-mediated reduction in the TRESK component may induce depolarization, increase excitability, and attenuate the calcium dependence of the membrane potential by reducing the calcineurin-activated fraction in the ensemble background K+ current.
Collapse
Affiliation(s)
- Enikő Pergel
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; (E.P.); (D.B.); (I.V.)
| | - Dániel J. Tóth
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; (E.P.); (D.B.); (I.V.)
- HUN-REN-SU Molecular Physiology Research Group, Hungarian Research Network and Semmelweis University, 1094 Budapest, Hungary
| | - Dóra Baukál
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; (E.P.); (D.B.); (I.V.)
| | - Irén Veres
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; (E.P.); (D.B.); (I.V.)
| | - Gábor Czirják
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary; (E.P.); (D.B.); (I.V.)
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Sun W, Yang F, Wang Y, Yang Y, Du R, Wang X, Luo Z, Wu J, Chen J. Sortilin-Mediated Inhibition of TREK1/2 Channels in Primary Sensory Neurons Promotes Prediabetic Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310295. [PMID: 38626370 PMCID: PMC11187941 DOI: 10.1002/advs.202310295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/29/2024] [Indexed: 04/18/2024]
Abstract
Neuropathic pain can occur during the prediabetic stage, even in the absence of hyperglycemia. The presence of prediabetic neuropathic pain (PDNP) poses challenges to the management of individuals with prediabetes. However, the mechanisms underlying this pain remain unclear. This study aims to investigate the underlying mechanism and identify potential therapeutic targets of PDNP. A prediabetic animal model induced by a high-energy diet exhibits both mechanical allodynia and thermal hyperalgesia. Furthermore, hyperexcitability and decreased potassium currents are observed in the dorsal root ganglion (DRG) neurons of these rats. TREK1 and TREK2 channels, which belong to the two-pore-domain K+ channel (K2P) family and play an important role in controlling cellular excitability, are downregulated in DRG neurons. Moreover, this alteration is modulated by Sortilin, a molecular partner that modulates the expression of TREK1. The overexpression of Sortilin negatively affects the expression of TREK1 and TREK2, leading to increased neuronal excitability in the DRG and enhanced peripheral pain sensitivity in rats. Moreover, the downregulation of Sortilin or activation of TREK1 and TREK2 channels by genetic or pharmacological approaches can alleviate PDNP. Therefore, targeting the Sortilin-mediated TREK1/2 pathway may provide a therapeutic approach for ameliorating PDNP.
Collapse
Affiliation(s)
- Wei Sun
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Fan Yang
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Yan Wang
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Yan Yang
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Rui Du
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Xiao‐Liang Wang
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Zhi‐Xin Luo
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
| | - Jun‐Jie Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of Orthodontics, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi Province710032P. R. China
| | - Jun Chen
- Institute for Biomedical Sciences of PainTangdu HospitalThe Fourth Military Medical UniversityXi'anShaanxi Province710038P. R. China
- Present address:
Sanhang Institute for Brain Science and Technology (SiBST)School of Medical Research, Northwestern Polytechnical University (NPU)Xi'an Shaanxi710129P. R. China
| |
Collapse
|
12
|
Zalaquett NG, Salameh E, Kim JM, Ghanbarian E, Tawk K, Abouzari M. The Dawn and Advancement of the Knowledge of the Genetics of Migraine. J Clin Med 2024; 13:2701. [PMID: 38731230 PMCID: PMC11084801 DOI: 10.3390/jcm13092701] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Migraine is a prevalent episodic brain disorder known for recurrent attacks of unilateral headaches, accompanied by complaints of photophobia, phonophobia, nausea, and vomiting. Two main categories of migraine are migraine with aura (MA) and migraine without aura (MO). Main body: Early twin and population studies have shown a genetic basis for these disorders, and efforts have been invested since to discern the genes involved. Many techniques, including candidate-gene association studies, loci linkage studies, genome-wide association, and transcription studies, have been used for this goal. As a result, several genes were pinned with concurrent and conflicting data among studies. It is important to understand the evolution of techniques and their findings. Conclusions: This review provides a chronological understanding of the different techniques used from the dawn of migraine genetic investigations and the genes linked with the migraine subtypes.
Collapse
Affiliation(s)
- Nader G. Zalaquett
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Elio Salameh
- Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Jonathan M. Kim
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Elham Ghanbarian
- Department of Neurology, University of California, Irvine, CA 92617, USA
| | - Karen Tawk
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Mehdi Abouzari
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Sutherland HG, Jenkins B, Griffiths LR. Genetics of migraine: complexity, implications, and potential clinical applications. Lancet Neurol 2024; 23:429-446. [PMID: 38508838 DOI: 10.1016/s1474-4422(24)00026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 03/22/2024]
Abstract
Migraine is a common neurological disorder with large burden in terms of disability for individuals and costs for society. Accurate diagnosis and effective treatments remain priorities. Understanding the genetic factors that contribute to migraine risk and symptom manifestation could improve individual management. Migraine has a strong genetic basis that includes both monogenic and polygenic forms. Some distinct, rare, familial migraine subtypes are caused by pathogenic variants in genes involved in ion transport and neurotransmitter release, suggesting an underlying vulnerability of the excitatory-inhibitory balance in the brain, which might be exacerbated by disruption of homoeostasis and lead to migraine. For more prevalent migraine subtypes, genetic studies have identified many susceptibility loci, implicating genes involved in both neuronal and vascular pathways. Genetic factors can also reveal the nature of relationships between migraine and its associated biomarkers and comorbidities and could potentially be used to identify new therapeutic targets and predict treatment response.
Collapse
Affiliation(s)
- Heidi G Sutherland
- Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Bronwyn Jenkins
- Department of Neurology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Lyn R Griffiths
- Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Suryavanshi P, Sawant-Pokam P, Clair S, Brennan KC. Increased presynaptic excitability in a migraine with aura mutation. Brain 2024; 147:680-697. [PMID: 37831655 PMCID: PMC10834252 DOI: 10.1093/brain/awad326] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 10/15/2023] Open
Abstract
Migraine is a common and disabling neurological disorder. The headache and sensory amplifications of migraine are attributed to hyperexcitable sensory circuits, but a detailed understanding remains elusive. A mutation in casein kinase 1 delta (CK1δ) was identified in non-hemiplegic familial migraine with aura and advanced sleep phase syndrome. Mice carrying the CK1δT44A mutation were more susceptible to spreading depolarization (the phenomenon that underlies migraine aura), but mechanisms underlying this migraine-relevant phenotype were not known. We used a combination of whole-cell electrophysiology and multiphoton imaging, in vivo and in brain slices, to compare CK1δT44A mice (adult males) to their wild-type littermates. We found that despite comparable synaptic activity at rest, CK1δT44A neurons were more excitable upon repetitive stimulation than wild-type, with a reduction in presynaptic adaptation at excitatory but not inhibitory synapses. The mechanism of this adaptation deficit was a calcium-dependent enhancement of the size of the readily releasable pool of synaptic vesicles, and a resultant increase in glutamate release, in CK1δT44A compared to wild-type synapses. Consistent with this mechanism, CK1δT44A neurons showed an increase in the cumulative amplitude of excitatory post-synaptic currents, and a higher excitation-to-inhibition ratio during sustained activity compared to wild-type. At a local circuit level, action potential bursts elicited in CK1δT44A neurons triggered an increase in recurrent excitation compared to wild-type, and at a network level, CK1δT44A mice showed a longer duration of 'up state' activity, which is dependent on recurrent excitation. Finally, we demonstrated that the spreading depolarization susceptibility of CK1δT44A mice could be returned to wild-type levels with the same intervention (reduced extracellular calcium) that normalized presynaptic adaptation. Taken together, these findings show a stimulus-dependent presynaptic gain of function at glutamatergic synapses in a genetic model of migraine, that accounts for the increased spreading depolarization susceptibility and may also explain the sensory amplifications that are associated with the disease.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Interdepartmental Neuroscience Program, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA
| | - Punam Sawant-Pokam
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Sarah Clair
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - K C Brennan
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
15
|
Della Pietra A, Gómez Dabó L, Mikulenka P, Espinoza-Vinces C, Vuralli D, Baytekin I, Martelletti P, Giniatullin R. Mechanosensitive receptors in migraine: a systematic review. J Headache Pain 2024; 25:6. [PMID: 38221631 PMCID: PMC10788982 DOI: 10.1186/s10194-023-01710-1] [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: 11/14/2023] [Accepted: 12/25/2023] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND Migraine is a debilitating neurological disorder with pain profile, suggesting exaggerated mechanosensation. Mechanosensitive receptors of different families, which specifically respond to various mechanical stimuli, have gathered increasing attention due to their potential role in migraine related nociception. Understanding these mechanisms is of principal importance for improved therapeutic strategies. This systematic review comprehensively examines the involvement of mechanosensitive mechanisms in migraine pain pathways. METHODS A systematic search across the Cochrane Library, Scopus, Web of Science, and Medline was conducted on 8th August 2023 for the period from 2000 to 2023, according to PRISMA guidelines. The review was constructed following a meticulous evaluation by two authors who independently applied rigorous inclusion criteria and quality assessments to the selected studies, upon which all authors collectively wrote the review. RESULTS We identified 36 relevant studies with our analysis. Additionally, 3 more studies were selected by literature search. The 39 papers included in this systematic review cover the role of the putative mechanosensitive Piezo and K2P, as well as ASICs, NMDA, and TRP family of channels in the migraine pain cascade. The outcome of the available knowledge, including mainly preclinical animal models of migraine and few clinical studies, underscores the intricate relationship between mechanosensitive receptors and migraine pain symptoms. The review presents the mechanisms of activation of mechanosensitive receptors that may be involved in the generation of nociceptive signals and migraine associated clinical symptoms. The gender differences of targeting these receptors as potential therapeutic interventions are also acknowledged as well as the challenges related to respective drug development. CONCLUSIONS Overall, this analysis identified key molecular players and uncovered significant gaps in our understanding of mechanotransduction in migraine. This review offers a foundation for filling these gaps and suggests novel therapeutic options for migraine treatments based on achievements in the emerging field of mechano-neurobiology.
Collapse
Affiliation(s)
- Adriana Della Pietra
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Laura Gómez Dabó
- Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Petr Mikulenka
- Department of Neurology, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | | | - Doga Vuralli
- Department of Neurology and Algology, Neuroscience and Neurotechnology Center of Excellence, Neuropsychiatry Center, Gazi University, Faculty of Medicine, Ankara, Turkey
| | - Isil Baytekin
- Department of Neurology, Bakirkoy Research and Training Hospital for Psychiatry, Neurology and Neurosurgery, Istanbul, Turkey
| | - Paolo Martelletti
- School of Health Sciences, Unitelma Sapienza University of Rome, Rome, Italy
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
16
|
Spekker E, Nagy-Grócz G, Vécsei L. Ion Channel Disturbances in Migraine Headache: Exploring the Potential Role of the Kynurenine System in the Context of the Trigeminovascular System. Int J Mol Sci 2023; 24:16574. [PMID: 38068897 PMCID: PMC10706278 DOI: 10.3390/ijms242316574] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Migraine is a primary headache disorder, which is an enormous burden to the healthcare system. While some aspects of the pathomechanism of migraines remain unknown, the most accepted theory is that activation and sensitization of the trigeminovascular system are essential during migraine attacks. In recent decades, it has been suggested that ion channels may be important participants in the pathogenesis of migraine. Numerous ion channels are expressed in the peripheral and central nervous systems, including the trigeminovascular system, affecting neuron excitability, synaptic energy homeostasis, inflammatory signaling, and pain sensation. Dysfunction of ion channels could result in neuronal excitability and peripheral or central sensitization. This narrative review covers the current understanding of the biological mechanisms leading to activation and sensitization of the trigeminovascular pain pathway, with a focus on recent findings on ion channel activation and modulation. Furthermore, we focus on the kynurenine pathway since this system contains kynurenic acid, which is an endogenous glutamate receptor antagonist substance, and it has a role in migraine pathophysiology.
Collapse
Affiliation(s)
| | - Gábor Nagy-Grócz
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, H-6726 Szeged, Hungary
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6725 Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, H-6725 Szeged, Hungary;
- HUN-REN-SZTE Neuroscience Research Group, University of Szeged, H-6725 Szeged, Hungary
| |
Collapse
|
17
|
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
|
18
|
Bogdanović I, Opačić M, Baščarević V, Raičević S, Ilić R, Grujičić D, Spasojević I, Ristić AJ. A potential role of mechano-gated potassium channels in meningioma-related seizures. Heliyon 2023; 9:e20761. [PMID: 37860528 PMCID: PMC10582377 DOI: 10.1016/j.heliyon.2023.e20761] [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: 06/08/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
Every third patient with intracranial meningioma develops seizures of poorly understood etiology. Tumor and peritumoral edema may exert mechanical pressure on the cortex that may affect mechano-gated potassium channels - KCNK2 and KCNK4. These channels regulate neuron excitability and have been related to seizures in some other conditions. The objective of the present study was to explore a potential relation between the levels of these proteins in tumor tissue and adjacent cortex and seizures development. The study included 19 meningioma patients that presented one or more preoperative seizures and 24 patients with no seizures. Tissue samples were collected in the course of surgical removal of the meningioma. Postoperative seizure freedom was achieved in 11 out of 19 patients. The relative level of KCNK2 in the cortical tissue was lower in patients with preoperative seizures. On the other hand, cortical tissue level of KCNK4 was higher in patients that became seizure-free after the surgery. In addition, relative levels of KCNK4 in the cortical and tumor tissue appear to be lowered by the treatment with anti-seizure medication levetiracetam. These results imply that KCNK2 and KCNK4 may be involved in the development of meningioma-related seizures and may represent promising therapeutic targets.
Collapse
Affiliation(s)
- Ivan Bogdanović
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Miloš Opačić
- University of Belgrade - Institute for Multidisciplinary Research, Life Sciences Department, Kneza Višeslava 1, 11030, Belgrade, Serbia
| | - Vladimir Baščarević
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Savo Raičević
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Rosanda Ilić
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Danica Grujičić
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovića 4, 11000, Belgrade, Serbia
| | - Ivan Spasojević
- University of Belgrade - Institute for Multidisciplinary Research, Life Sciences Department, Kneza Višeslava 1, 11030, Belgrade, Serbia
| | - Aleksandar J. Ristić
- Center for Epilepsy and Sleep Disorders, Neurology Clinic, Clinical Center of Serbia, Dr Subotića Starijeg 6, 11000, Belgrade, Serbia
| |
Collapse
|
19
|
Barry AM, Zhao N, Yang X, Bennett DL, Baskozos G. Deep RNA-seq of male and female murine sensory neuron subtypes after nerve injury. Pain 2023; 164:2196-2215. [PMID: 37318015 PMCID: PMC10502896 DOI: 10.1097/j.pain.0000000000002934] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/27/2023] [Accepted: 02/05/2023] [Indexed: 06/16/2023]
Abstract
ABSTRACT Dorsal root ganglia (DRG) neurons have been well described for their role in driving both acute and chronic pain. Although nerve injury is known to cause transcriptional dysregulation, how this differs across neuronal subtypes and the impact of sex is unclear. Here, we study the deep transcriptional profiles of multiple murine DRG populations in early and late pain states while considering sex. We have exploited currently available transgenics to label numerous subpopulations for fluorescent-activated cell sorting and subsequent transcriptomic analysis. Using bulk tissue samples, we are able to circumvent the issues of low transcript coverage and drop-outs seen with single-cell data sets. This increases our power to detect novel and even subtle changes in gene expression within neuronal subtypes and discuss sexual dimorphism at the neuronal subtype level. We have curated this resource into an accessible database for other researchers ( https://livedataoxford.shinyapps.io/drg-directory/ ). We see both stereotyped and unique subtype signatures in injured states after nerve injury at both an early and late timepoint. Although all populations contribute to a general injury signature, subtype enrichment changes can also be seen. Within populations, there is not a strong intersection of sex and injury, but previously unknown sex differences in naïve states-particularly in Aβ-RA + Aδ-low threshold mechanoreceptors-still contribute to differences in injured neurons.
Collapse
Affiliation(s)
- Allison M. Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Na Zhao
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Georgios Baskozos
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Pleș H, Florian IA, Timis TL, Covache-Busuioc RA, Glavan LA, Dumitrascu DI, Popa AA, Bordeianu A, Ciurea AV. Migraine: Advances in the Pathogenesis and Treatment. Neurol Int 2023; 15:1052-1105. [PMID: 37755358 PMCID: PMC10535528 DOI: 10.3390/neurolint15030067] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
This article presents a comprehensive review on migraine, a prevalent neurological disorder characterized by chronic headaches, by focusing on their pathogenesis and treatment advances. By examining molecular markers and leveraging imaging techniques, the research identifies key mechanisms and triggers in migraine pathology, thereby improving our understanding of its pathophysiology. Special emphasis is given to the role of calcitonin gene-related peptide (CGRP) in migraine development. CGRP not only contributes to symptoms but also represents a promising therapeutic target, with inhibitors showing effectiveness in migraine management. The article further explores traditional medical treatments, scrutinizing the mechanisms, benefits, and limitations of commonly prescribed medications. This provides a segue into an analysis of emerging therapeutic strategies and their potential to enhance migraine management. Finally, the paper delves into neuromodulation as an innovative treatment modality. Clinical studies indicating its effectiveness in migraine management are reviewed, and the advantages and limitations of this technique are discussed. In summary, the article aims to enhance the understanding of migraine pathogenesis and present novel therapeutic possibilities that could revolutionize patient care.
Collapse
Affiliation(s)
- Horia Pleș
- Department of Neurosurgery, Centre for Cognitive Research in Neuropsychiatric Pathology (NeuroPsy-Cog), “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Ioan-Alexandru Florian
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Teodora-Larisa Timis
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Razvan-Adrian Covache-Busuioc
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| | - Luca-Andrei Glavan
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| | - David-Ioan Dumitrascu
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| | - Andrei Adrian Popa
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| | - Andrei Bordeianu
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Neurosurgery Department, “Carol Davila” University of Medicine and Pharmacy, 020021 București, Romania; (R.-A.C.-B.); (L.-A.G.); (D.-I.D.); (A.A.P.); (A.B.); (A.V.C.)
| |
Collapse
|
21
|
Gosalia H, Karsan N, Goadsby PJ. Genetic Mechanisms of Migraine: Insights from Monogenic Migraine Mutations. Int J Mol Sci 2023; 24:12697. [PMID: 37628876 PMCID: PMC10454024 DOI: 10.3390/ijms241612697] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Migraine is a disabling neurological disorder burdening patients globally. Through the increasing development of preclinical and clinical experimental migraine models, advancing appreciation of the extended clinical phenotype, and functional neuroimaging studies, we can further our understanding of the neurobiological basis of this highly disabling condition. Despite increasing understanding of the molecular and chemical architecture of migraine mechanisms, many areas require further investigation. Research over the last three decades has suggested that migraine has a strong genetic basis, based on the positive family history in most patients, and this has steered exploration into possibly implicated genes. In recent times, human genome-wide association studies and rodent genetic migraine models have facilitated our understanding, but most migraine seems polygenic, with the monogenic migraine mutations being considerably rarer, so further large-scale studies are required to elucidate fully the genetic underpinnings of migraine and the translation of these to clinical practice. The monogenic migraine mutations cause severe aura phenotypes, amongst other symptoms, and offer valuable insights into the biology of aura and the relationship between migraine and other conditions, such as vascular disease and sleep disorders. This review will provide an outlook of what is known about some monogenic migraine mutations, including familial hemiplegic migraine, familial advanced sleep-phase syndrome, and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy.
Collapse
Affiliation(s)
- Helin Gosalia
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
| | - Nazia Karsan
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
| | - Peter J. Goadsby
- Headache Group, The Wolfson Sensory, Pain and Rehabilitation Centre, NIHR King’s Clinical Research Facility, & SLaM Biomedical Research Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9PJ, UK; (H.G.); (N.K.)
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
22
|
Al-Hassany L, Boucherie DM, Creeney H, van Drie RWA, Farham F, Favaretto S, Gollion C, Grangeon L, Lyons H, Marschollek K, Onan D, Pensato U, Stanyer E, Waliszewska-Prosół M, Wiels W, Chen HZ, Amin FM. Future targets for migraine treatment beyond CGRP. J Headache Pain 2023; 24:76. [PMID: 37370051 DOI: 10.1186/s10194-023-01567-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/14/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Migraine is a disabling and chronic neurovascular headache disorder. Trigeminal vascular activation and release of calcitonin gene-related peptide (CGRP) play a pivotal role in the pathogenesis of migraine. This knowledge has led to the development of CGRP(-receptor) therapies. Yet, a substantial proportion of patients do not respond to these treatments. Therefore, alternative targets for future therapies are warranted. The current narrative review provides a comprehensive overview of the pathophysiological role of these possible non-CGRP targets in migraine. FINDINGS We covered targets of the metabotropic receptors (pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), amylin, and adrenomedullin), intracellular targets (nitric oxide (NO), phosphodiesterase-3 (PDE3) and -5 (PDE5)), and ion channels (potassium, calcium, transient receptor potential (TRP), and acid-sensing ion channels (ASIC)). The majority of non-CGRP targets were able to induce migraine-like attacks, except for (i) calcium channels, as it is not yet possible to directly target channels to elucidate their precise involvement in migraine; (ii) TRP channels, activation of which can induce non-migraine headache; and (iii) ASICs, as their potential in inducing migraine attacks has not been investigated thus far. Drugs that target its receptors exist for PACAP, NO, and the potassium, TRP, and ASIC channels. No selective drugs exist for the other targets, however, some existing (migraine) treatments appear to indirectly antagonize responses to amylin, adrenomedullin, and calcium channels. Drugs against PACAP, NO, potassium channels, TRP channels, and only a PAC1 antibody have been tested for migraine treatment, albeit with ambiguous results. CONCLUSION While current research on these non-CGRP drug targets has not yet led to the development of efficacious therapies, human provocation studies using these targets have provided valuable insight into underlying mechanisms of migraine headaches and auras. Further studies are needed on these alternative therapies in non-responders of CGRP(-receptor) targeted therapies with the ultimate aim to pave the way towards a headache-free future for all migraine patients.
Collapse
Affiliation(s)
- Linda Al-Hassany
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Deirdre M Boucherie
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Hannah Creeney
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Ruben W A van Drie
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Cardiology, Division of Experimental Cardiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Fatemeh Farham
- Department of Headache, Iranian Centre of Neurological Researchers, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Silvia Favaretto
- Headache Center, Neurology Clinic, University Hospital of Padua, Padua, Italy
| | - Cédric Gollion
- Department of Neurology, University Hospital of Toulouse, Toulouse, France
| | - Lou Grangeon
- Neurology Department, Rouen University Hospital, Rouen, France
| | - Hannah Lyons
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Karol Marschollek
- Department of Neurology, Wroclaw Medical University, Wrocław, Poland
| | - Dilara Onan
- Spine Health Unit, Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Umberto Pensato
- Neurology and Stroke Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Pieve Emanuele, Milan, Italy
| | - Emily Stanyer
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | | | - Wietse Wiels
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hui Zhou Chen
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Faisal Mohammad Amin
- Danish Headache Center, Department of Neurology, Faculty of Health and Medical Sciences, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark.
- Department of Neurorehabilitation/Traumatic Brain Injury, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Kim SS, Park J, Kim E, Hwang EM, Park JY. β-COP Suppresses the Surface Expression of the TREK2. Cells 2023; 12:1500. [PMID: 37296621 PMCID: PMC10252889 DOI: 10.3390/cells12111500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
K2P channels, also known as two-pore domain K+ channels, play a crucial role in maintaining the cell membrane potential and contributing to potassium homeostasis due to their leaky nature. The TREK, or tandem of pore domains in a weak inward rectifying K+ channel (TWIK)-related K+ channel, subfamily within the K2P family consists of mechanical channels regulated by various stimuli and binding proteins. Although TREK1 and TREK2 within the TREK subfamily share many similarities, β-COP, which was previously known to bind to TREK1, exhibits a distinct binding pattern to other members of the TREK subfamily, including TREK2 and the TRAAK (TWIK-related acid-arachidonic activated K+ channel). In contrast to TREK1, β-COP binds to the C-terminus of TREK2 and reduces its cell surface expression but does not bind to TRAAK. Furthermore, β-COP cannot bind to TREK2 mutants with deletions or point mutations in the C-terminus and does not affect the surface expression of these TREK2 mutants. These results emphasize the unique role of β-COP in regulating the surface expression of the TREK family.
Collapse
Affiliation(s)
- Seong-Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
| | - Jimin Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Republic of Korea
| | - Eunju Kim
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea;
| | - Eun Mi Hwang
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea;
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Republic of Korea
- ASTRION, Inc., Seoul 02842, Republic of Korea
| |
Collapse
|
24
|
Debreczeni D, Baukál D, Pergel E, Veres I, Czirják G. Critical contribution of the intracellular C-terminal region to TRESK channel activity is revealed by the epithelial Na + current ratio (ENaR) method. J Biol Chem 2023; 299:104737. [PMID: 37084812 DOI: 10.1016/j.jbc.2023.104737] [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: 01/23/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
TRESK (K2P18.1) possesses unique structural proportions within the K2P background potassium channel family. The previously described TRESK regulatory mechanisms are based on the long intracellular loop between the second and third transmembrane segments (TMS). However, the functional significance of the exceptionally short intracellular C-terminal region (iCtr) following the fourth TMS has not yet been examined. In the present study, we investigated TRESK constructs modified at the iCtr by two-electrode voltage clamp and the newly developed epithelial sodium current ratio (ENaR) method in Xenopus oocytes. The ENaR method allowed the evaluation of channel activity by exclusively using electrophysiology, and provided data that are otherwise not readily available under whole-cell conditions. TRESK homodimer was connected with two ENaC (epithelial Na+ channel) heterotrimers and the Na+ current was measured as an internal reference, proportional to the number of channels in the plasma membrane. Modifications of TRESK iCtr resulted in diverse functional effects, indicating a complex contribution of this region to K+ channel activity. Mutations of positive residues in proximal iCtr locked TRESK in a low activity, calcineurin-insensitive state, although this phosphatase binds to distant motifs in the loop region. Accordingly, mutations in proximal iCtr may prevent the transmission of modulation to the gating machinery. Replacing distal iCtr with a sequence designed to interact with the inner surface of the plasma membrane increased the activity of the channel to unprecedented levels, as indicated by ENaR and single channel measurements. In conclusion, the distal iCtr is a major positive determinant of TRESK function.
Collapse
Affiliation(s)
| | - Dóra Baukál
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Enikő Pergel
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Irén Veres
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gábor Czirják
- Department of Physiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
25
|
A photoswitchable inhibitor of TREK channels controls pain in wild-type intact freely moving animals. Nat Commun 2023; 14:1160. [PMID: 36859433 PMCID: PMC9977718 DOI: 10.1038/s41467-023-36806-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
By endowing light control of neuronal activity, optogenetics and photopharmacology are powerful methods notably used to probe the transmission of pain signals. However, costs, animal handling and ethical issues have reduced their dissemination and routine use. Here we report LAKI (Light Activated K+ channel Inhibitor), a specific photoswitchable inhibitor of the pain-related two-pore-domain potassium TREK and TRESK channels. In the dark or ambient light, LAKI is inactive. However, alternating transdermal illumination at 365 nm and 480 nm reversibly blocks and unblocks TREK/TRESK current in nociceptors, enabling rapid control of pain and nociception in intact and freely moving mice and nematode. These results demonstrate, in vivo, the subcellular localization of TREK/TRESK at the nociceptor free nerve endings in which their acute inhibition is sufficient to induce pain, showing LAKI potential as a valuable tool for TREK/TRESK channel studies. More importantly, LAKI gives the ability to reversibly remote-control pain in a non-invasive and physiological manner in naive animals, which has utility in basic and translational pain research but also in in vivo analgesic drug screening and validation, without the need of genetic manipulations or viral infection.
Collapse
|
26
|
Grangeon L, Lange KS, Waliszewska-Prosół M, Onan D, Marschollek K, Wiels W, Mikulenka P, Farham F, Gollion C, Ducros A, on behalf of the European Headache Federation School of Advanced Studies (EHF-SAS). Genetics of migraine: where are we now? J Headache Pain 2023; 24:12. [PMID: 36800925 PMCID: PMC9940421 DOI: 10.1186/s10194-023-01547-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/07/2023] [Indexed: 02/21/2023] Open
Abstract
Migraine is a complex brain disorder explained by the interaction of genetic and environmental factors. In monogenic migraines, including familial hemiplegic migraine and migraine with aura associated with hereditary small-vessel disorders, the identified genes code for proteins expressed in neurons, glial cells, or vessels, all of which increase susceptibility to cortical spreading depression. The study of monogenic migraines has shown that the neurovascular unit plays a prominent role in migraine. Genome-wide association studies have identified numerous susceptibility variants that each result in only a small increase in overall migraine risk. The more than 180 known variants belong to several complex networks of "pro-migraine" molecular abnormalities, which are mainly neuronal or vascular. Genetics has also highlighted the importance of shared genetic factors between migraine and its major co-morbidities, including depression and high blood pressure. Further studies are still needed to map all of the susceptibility loci for migraine and then to understand how these genomic variants lead to migraine cell phenotypes.
Collapse
Affiliation(s)
- Lou Grangeon
- grid.41724.340000 0001 2296 5231Neurology Department, CHU de Rouen, Rouen, France
| | - Kristin Sophie Lange
- grid.6363.00000 0001 2218 4662Neurology Department, Charité – Universitätsmedizin Berlin, Berlin, Germany ,grid.6363.00000 0001 2218 4662Center for Stroke Research Berlin (CSB), Charité – Universitätsmedizin, Berlin, Germany
| | - Marta Waliszewska-Prosół
- grid.4495.c0000 0001 1090 049XDepartment of Neurology, Wrocław Medical University, Wrocław, Poland
| | - Dilara Onan
- grid.14442.370000 0001 2342 7339Hacettepe University, Faculty of Physical Therapy and Rehabilitation, Ankara, Turkey
| | - Karol Marschollek
- grid.4495.c0000 0001 1090 049XDepartment of Neurology, Wrocław Medical University, Wrocław, Poland
| | - Wietse Wiels
- grid.8767.e0000 0001 2290 8069Department of Neurology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Petr Mikulenka
- grid.412819.70000 0004 0611 1895Department of Neurology, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Fatemeh Farham
- grid.411705.60000 0001 0166 0922Headache Department, Iranian Centre of Neurological Researchers, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Cédric Gollion
- grid.411175.70000 0001 1457 2980Neurology Department, CHU de Toulouse, Toulouse, France
| | - Anne Ducros
- Neurology Department, CHU de Montpellier, 80 avenue Augustin Fliche, 34295, Montpellier, France.
| | | |
Collapse
|
27
|
Sanchez Del Rio M, Cutrer FM. Pathophysiology of migraine aura. HANDBOOK OF CLINICAL NEUROLOGY 2023; 198:71-83. [PMID: 38043972 DOI: 10.1016/b978-0-12-823356-6.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Migraine aura occurs in about a third of patients with migraine and consists of a group of transient focal neurological symptoms that appear from 5 to 60min and then resolve prior to or in the early phase of a migraine headache attack. Migraine auras may consist of visual, language, unilateral sensory, or motor symptoms. There has been considerable debate as to the origins of the migrainous aura. Investigations during physiologically induced visual auras suggest that the phenomenon of cortical spreading depression or its human equivalent underpins the migraine aura. Single gene defects have been linked to relatively rare forms of the motor subtypes of aura known as familial hemiplegic migraine (FHM). These include CACNA1A (FHM1), ATP1A2 (FHM2), and SCN1A (FHM3). In the familial hemiplegic forms of migraine, the more typical forms of aura are almost always also present. Despite ample epidemiological evidence of increased heritability of migraine with aura compared to migraine without aura, identification of the specific variants driving susceptibility to the more common forms of aura has been problematic thus far. In the first genome-wide association study (GWAS) that focused migraine with aura, a single SNP rs835740 reached genome-wide significance. Unfortunately, the SNP did show statistical significance in a later meta-analysis which included GWAS data from subsequent studies. Here, we review the clinical features, pathophysiological theories, and currently available potential evidence for the genetic basis of migraine aura.
Collapse
|
28
|
Markel KA, Curtis D. Study of variants in genes implicated in rare familial migraine syndromes and their association with migraine in 200,000 exome-sequenced UK Biobank participants. Ann Hum Genet 2022; 86:353-360. [PMID: 36044383 DOI: 10.1111/ahg.12484] [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: 03/18/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND A number of genes have been implicated in rare familial syndromes which have migraine as part of their phenotype but these genes have not previously been implicated in the common form of migraine. METHODS Among exome-sequenced participants in the UK Biobank, we identified 7194 migraine cases with the remaining 193,433 participants classified as controls. We investigated rare variants in 10 genes previously reported to be implicated in conditions with migraine as a prominent part of the phenotype and carried out gene- and variant-based tests for association. RESULTS We found no evidence for association of these genes or variants with the common form of migraine seen in our subjects. In particular, a frameshift variant in KCNK18, p.(Phe139Trpfs*24), which had been shown to segregate with migraine with aura in a multiply affected pedigree, was found in 196 (0.10%) controls as well as in 10 (0.14%) cases (χ2 = 0.96, 1 df, p = 0.33). CONCLUSIONS Since there is no other reported evidence to implicate KCNK18, we conclude that this gene and its product, TRESK, should no longer be regarded as being involved in migraine aetiology. Overall, we do not find that rare, functional variants in genes previously implicated to be involved in familial syndromes including migraine as part of the phenotype make a contribution to the commoner forms of migraine observed in this population.
Collapse
Affiliation(s)
| | - David Curtis
- UCL Genetics Institute, University College London, London, UK.,Centre for Psychiatry, Queen Mary University of London, London, UK
| |
Collapse
|
29
|
Benarroch E. What Is the Role of 2-Pore Domain Potassium Channels (K2P) in Pain? Neurology 2022; 99:516-521. [PMID: 36123135 DOI: 10.1212/wnl.0000000000201197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 11/15/2022] Open
|
30
|
Khoubza L, Gilbert N, Kim EJ, Chatelain FC, Feliciangeli S, Abelanet S, Kang D, Lesage F, Bichet D. Alkaline-sensitive two-pore domain potassium channels form functional heteromers in pancreatic β-cells. J Biol Chem 2022; 298:102447. [PMID: 36063992 PMCID: PMC9520024 DOI: 10.1016/j.jbc.2022.102447] [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: 11/08/2021] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022] Open
Abstract
Two-pore domain K+ channels (K2P channels), active as dimers, produce inhibitory currents regulated by a variety of stimuli. Among them, TWIK1-related alkalinization-activated K+ channel 1 (TALK1), TWIK1-related alkalinization-activated K+ channel 2 (TALK2), and TWIK1-related acid-sensitive K+ channel 2 (TASK2) form a subfamily of structurally related K2P channels stimulated by extracellular alkalosis. The human genes encoding these proteins are clustered at chromosomal region 6p21 and coexpressed in multiple tissues, including the pancreas. The question whether these channels form functional heteromers remained open. By analyzing single-cell transcriptomic data, we show that these channels are coexpressed in insulin-secreting pancreatic β-cells. Using in situ proximity ligation assay and electrophysiology, we show that they form functional heterodimers both upon heterologous expression and under native conditions in human pancreatic β-cells. We demonstrate that heteromerization of TALK2 with TALK1 or with TASK2 endows TALK2 with sensitivity to extracellular alkalosis in the physiological range. We further show that the association of TASK2 with TALK1 and TALK2 increases their unitary conductance. These results provide a new example of heteromerization in the K2P channel family expanding the range of the potential physiological and pathophysiological roles of TALK1/TALK2/TASK2 channels, not only in insulin-secreting cells but also in the many other tissues in which they are coexpressed.
Collapse
Affiliation(s)
- Lamyaa Khoubza
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Nicolas Gilbert
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Eun-Jin Kim
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Franck C Chatelain
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Sylvain Feliciangeli
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France; Inserm, Paris, France
| | - Sophie Abelanet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| | - Dawon Kang
- Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University, Jinju, South Korea
| | - Florian Lesage
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France; Inserm, Paris, France.
| | - Delphine Bichet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, Valbonne, France
| |
Collapse
|
31
|
Ślęczkowska M, Almomani R, Marchi M, de Greef BTA, Sopacua M, Hoeijmakers JGJ, Lindsey P, Salvi E, Bönhof GJ, Ziegler D, Malik RA, Waxman SG, Lauria G, Faber CG, Smeets HJM, Gerrits MM. Peripheral Ion Channel Gene Screening in Painful- and Painless-Diabetic Neuropathy. Int J Mol Sci 2022; 23:ijms23137190. [PMID: 35806193 PMCID: PMC9266298 DOI: 10.3390/ijms23137190] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Neuropathic pain is common in diabetic peripheral neuropathy (DN), probably caused by pathogenic ion channel gene variants. Therefore, we performed molecular inversion probes-next generation sequencing of 5 transient receptor potential cation channels, 8 potassium channels and 2 calcium-activated chloride channel genes in 222 painful- and 304 painless-DN patients. Twelve painful-DN (5.4%) patients showed potentially pathogenic variants (five nonsense/frameshift, seven missense, one out-of-frame deletion) in ANO3 (n = 3), HCN1 (n = 1), KCNK18 (n = 2), TRPA1 (n = 3), TRPM8 (n = 3) and TRPV4 (n = 1) and fourteen painless-DN patients (4.6%-three nonsense/frameshift, nine missense, one out-of-frame deletion) in ANO1 (n = 1), KCNK18 (n = 3), KCNQ3 (n = 1), TRPA1 (n = 2), TRPM8 (n = 1), TRPV1 (n = 3) and TRPV4 (n = 3). Missense variants were present in both conditions, presumably with loss- or gain-of-functions. KCNK18 nonsense/frameshift variants were found in painless/painful-DN, making a causal role in pain less likely. Surprisingly, premature stop-codons with likely nonsense-mediated RNA-decay were more frequent in painful-DN. Although limited in number, painful-DN patients with ion channel gene variants reported higher maximal pain during the night and day. Moreover, painful-DN patients with TRP variants had abnormal thermal thresholds and more severe pain during the night and day. Our results suggest a role of ion channel gene variants in neuropathic pain, but functional validation is required.
Collapse
Affiliation(s)
- Milena Ślęczkowska
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.A.); (P.L.); (H.J.M.S.)
- School of Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| | - Rowida Almomani
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.A.); (P.L.); (H.J.M.S.)
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.T.A.d.G.); (M.S.); (J.G.J.H.); (C.G.F.)
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy; (M.M.); (E.S.); (G.L.)
| | - Bianca T. A. de Greef
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.T.A.d.G.); (M.S.); (J.G.J.H.); (C.G.F.)
| | - Maurice Sopacua
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.T.A.d.G.); (M.S.); (J.G.J.H.); (C.G.F.)
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.T.A.d.G.); (M.S.); (J.G.J.H.); (C.G.F.)
| | - Patrick Lindsey
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.A.); (P.L.); (H.J.M.S.)
| | - Erika Salvi
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy; (M.M.); (E.S.); (G.L.)
| | - Gidon J. Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (G.J.B.); (D.Z.)
- Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Dan Ziegler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (G.J.B.); (D.Z.)
| | - Rayaz A. Malik
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK;
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha P.O. Box 24144, Qatar
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA;
- Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT 06516, USA
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy; (M.M.); (E.S.); (G.L.)
| | - Catharina G. Faber
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands; (B.T.A.d.G.); (M.S.); (J.G.J.H.); (C.G.F.)
| | - Hubert J. M. Smeets
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands; (R.A.); (P.L.); (H.J.M.S.)
- School of Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Monique M. Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands;
| |
Collapse
|
32
|
Finno CJ, Chen Y, Park S, Lee JH, Perez-Flores MC, Choi J, Yamoah EN. Cisplatin Neurotoxicity Targets Specific Subpopulations and K + Channels in Tyrosine-Hydroxylase Positive Dorsal Root Ganglia Neurons. Front Cell Neurosci 2022; 16:853035. [PMID: 35586548 PMCID: PMC9108181 DOI: 10.3389/fncel.2022.853035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Among the features of cisplatin chemotherapy-induced peripheral neuropathy are chronic pain and innocuous mechanical hypersensitivity. The complete etiology of the latter remains unknown. Here, we show that cisplatin targets a heterogeneous population of tyrosine hydroxylase-positive (TH+) primary afferent dorsal root ganglion neurons (DRGNs) in mice, determined using single-cell transcriptome and electrophysiological analyses. TH+ DRGNs regulate innocuous mechanical sensation through C-low threshold mechanoreceptors. A differential assessment of wild-type and vitamin E deficient TH+ DRGNs revealed heterogeneity and specific functional phenotypes. The TH+ DRGNs comprise; fast-adapting eliciting one action potential (AP; 1-AP), moderately-adapting (≥2-APs), in responses to square-pulse current injection, and spontaneously active (SA). Cisplatin increased the input resistance and AP frequency but reduced the temporal coding feature of 1-AP and ≥2-APs neurons. By contrast, cisplatin has no measurable effect on the SA neurons. Vitamin E reduced the cisplatin-mediated increased excitability but did not improve the TH+ neuron temporal coding properties. Cisplatin mediates its effect by targeting outward K+ current, likely carried through K2P18.1 (Kcnk18), discovered through the differential transcriptome studies and heterologous expression. Studies show a potential new cellular target for chemotherapy-induced peripheral neuropathy and implicate the possible neuroprotective effects of vitamin E in cisplatin chemotherapy.
Collapse
Affiliation(s)
- Carrie J. Finno
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | | | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| |
Collapse
|
33
|
Advances in tethered photopharmacology for precise optical control of signaling proteins. Curr Opin Pharmacol 2022; 63:102196. [PMID: 35245800 DOI: 10.1016/j.coph.2022.102196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/25/2022]
Abstract
To overcome the limitations of traditional pharmacology, the field of photopharmacology has developed around the central concept of using light to endow drug action with spatiotemporal precision. Tethered photopharmacology, where a photoswitchable ligand is covalently attached to a target protein, offers a particularly high degree of spatiotemporal control, as well as the ability to genetically target drug action and limit effects to specific protein subtypes. In this review, we describe the core engineering concepts of tethered pharmacology and highlight recent advances in harnessing the power of tethered photopharmacology for an expanded palette of targets and conjugation modes using new, complementary strategies. We also discuss the various applications, including mechanistic studies from the molecular biophysical realm to in vivo studies in behaving animals, that demonstrate the power of tethered pharmacology.
Collapse
|
34
|
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: 1.7] [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
|
35
|
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: 14] [Impact Index Per Article: 4.7] [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
|
36
|
Regulation of Two-Pore-Domain Potassium TREK Channels and their Involvement in Pain Perception and Migraine. Neurosci Lett 2022; 773:136494. [DOI: 10.1016/j.neulet.2022.136494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
|
37
|
Comes N, Gasull X, Callejo G. Proton Sensing on the Ocular Surface: Implications in Eye Pain. Front Pharmacol 2021; 12:773871. [PMID: 34899333 PMCID: PMC8652213 DOI: 10.3389/fphar.2021.773871] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 01/15/2023] Open
Abstract
Protons reaching the eyeball from exogenous acidic substances or released from damaged cells during inflammation, immune cells, after tissue injury or during chronic ophthalmic conditions, activate or modulate ion channels present in sensory nerve fibers that innervate the ocular anterior surface. Their identification as well as their role during disease is critical for the understanding of sensory ocular pathophysiology. They are likely to mediate some of the discomfort sensations accompanying several ophthalmic formulations and may represent novel targets for the development of new therapeutics for ocular pathologies. Among the ion channels expressed in trigeminal nociceptors innervating the anterior surface of the eye (cornea and conjunctiva) and annex ocular structures (eyelids), members of the TRP and ASIC families play a critical role in ocular acidic pain. Low pH (pH 6) activates TRPV1, a polymodal ion channel also activated by heat, capsaicin and hyperosmolar conditions. ASIC1, ASIC3 and heteromeric ASIC1/ASIC3 channels present in ocular nerve terminals are activated at pH 7.2–6.5, inducing pain by moderate acidifications of the ocular surface. These channels, together with TRPA1, are involved in acute ocular pain, as well as in painful sensations during allergic keratoconjunctivitis or other ophthalmic conditions, as blocking or reducing channel expression ameliorates ocular pain. TRPV1, TRPA1 and other ion channels are also present in corneal and conjunctival cells, promoting inflammation of the ocular surface after injury. In addition to the above-mentioned ion channels, members of the K2P and P2X ion channel families are also expressed in trigeminal neurons, however, their role in ocular pain remains unclear to date. In this report, these and other ion channels and receptors involved in acid sensing during ocular pathologies and pain are reviewed.
Collapse
Affiliation(s)
- Núria Comes
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gerard Callejo
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
38
|
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: 18] [Impact Index Per Article: 4.5] [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
|
39
|
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: 1.0] [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
|
40
|
Structural and compositional diversity in the kainate receptor family. Cell Rep 2021; 37:109891. [PMID: 34706237 PMCID: PMC8581553 DOI: 10.1016/j.celrep.2021.109891] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/10/2021] [Accepted: 10/05/2021] [Indexed: 01/17/2023] Open
Abstract
The kainate receptors (KARs) are members of the ionotropic glutamate receptor family and assemble into tetramers from a pool of five subunit types (GluK1–5). Each subunit confers distinct functional properties to a receptor, but the compositional and stoichiometric diversity of KAR tetramers is not well understood. To address this, we first solve the structure of the GluK1 homomer, which enables a systematic assessment of structural compatibility among KAR subunits. Next, we analyze single-cell RNA sequencing data, which reveal extreme diversity in the combinations of two or more KAR subunits co-expressed within the same cell. We then investigate the composition of individual receptor complexes using single-molecule fluorescence techniques and find that di-heteromers assembled from GluK1, GluK2, or GluK3 can form with all possible stoichiometries, while GluK1/K5, GluK2/K5, and GluK3/K5 can form 3:1 or 2:2 complexes. Finally, using three-color single-molecule imaging, we discover that KARs can form tri- and tetra-heteromers. Selvakumar et al. use cryo-electron microscopy, single-cell RNA sequencing analysis, and single-molecule fluorescence techniques to investigate the stoichiometric and assembly diversity of kainate receptors (KARs). The work gives insight into KAR molecular diversity and expands the potential KAR subunit combinations to include a variety of di-, tri-, and tetra-heteromers.
Collapse
|
41
|
A review of migraine genetics: gathering genomic and transcriptomic factors. Hum Genet 2021; 141:1-14. [PMID: 34686893 DOI: 10.1007/s00439-021-02389-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/16/2021] [Indexed: 01/28/2023]
Abstract
Migraine is a common and complex neurologic disorder that affects approximately 15-18% of the general population. Although the cause of migraine is unknown, some genetic studies have focused on unravelling rare and common variants underlying the pathophysiological mechanisms of this disorder. This review covers the advances in the last decade on migraine genetics, throughout the history of genetic methodologies used, including recent application of next-generation sequencing techniques. A thorough review of the literature interweaves the genomic and transcriptomic factors that will allow a better understanding of the mechanisms underlying migraine pathophysiology, concluding with the clinical utility landscape of genetic information and future consideration to creating a new frontier toward advancing the field of personalized medicine.
Collapse
|
42
|
Ávalos Prado P, Landra-Willm A, Verkest C, Ribera A, Chassot AA, Baron A, Sandoz G. TREK channel activation suppresses migraine pain phenotype. iScience 2021; 24:102961. [PMID: 34458705 PMCID: PMC8379698 DOI: 10.1016/j.isci.2021.102961] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/20/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022] Open
Abstract
Activation and sensitization of trigeminal ganglia (TG) sensory neurons, leading to the release of pro-inflammatory peptides such as calcitonin gene-related peptide (CGRP), are likely a key component in migraine-related headache induction. Reducing TG neuron excitability represents therefore an attractive alternative strategy to relieve migraine pain. Here by using pharmacology and genetic invalidation ex vivo and in vivo, we demonstrate that activating TREK1 and TREK2 two-pore-domain potassium (K2P) channels inhibits TG neuronal firing sufficiently to fully reverse the migraine-like phenotype induced by NO-donors in rodents. Finally, targeting TREK is as efficient as treatment with CGRP antagonists, which represents one of the most effective migraine therapies. Altogether, our results demonstrate that inhibiting TG excitability by pharmacological activation of TREK channels should be considered as an alternative to the current migraine treatment.
Collapse
Affiliation(s)
- Pablo Ávalos Prado
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
| | - Arnaud Landra-Willm
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
| | - Clément Verkest
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
- Universite Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Aurore Ribera
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
| | - Anne-Amandine Chassot
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
| | - Anne Baron
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
- Universite Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Guillaume Sandoz
- Université Côte d’Azur, CNRS, INSERM, iBV, Nice, France
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
- Fédération Hospitalo-Universitaire InovPain, Cote d'Azur University, University Hospital Centre Nice, Nice, Provence-Alpes-Côte d'Azur, France
- Corresponding author
| |
Collapse
|
43
|
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: 18] [Impact Index Per Article: 4.5] [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
|
44
|
Kanda H, Tonomura S, Gu JG. Effects of Cooling Temperatures via Thermal K2P Channels on Regeneration of High-Frequency Action Potentials at Nodes of Ranvier of Rat Aβ-Afferent Nerves. eNeuro 2021; 8:ENEURO.0308-21.2021. [PMID: 34462308 PMCID: PMC8445039 DOI: 10.1523/eneuro.0308-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 12/02/2022] Open
Abstract
Temperature-sensitive two-pore domain potassium channels (thermal K2P) are recently shown to cluster at nodes of Ranvier (NRs) and play a key role in action potential (AP) regeneration and conduction on Aβ-afferent nerves. Cooling temperatures affect AP regeneration and conduction on Aβ-afferent nerves but the underlying mechanisms are not completely understood. Here, we have performed patch-clamp recordings directly at the NR in an ex vivo trigeminal nerve preparation. We have characterized the effects of cooling temperatures on intrinsic electrophysiological properties and AP regeneration at the NR on rat Aβ-afferent nerves, and determined whether and how thermal K2P channels may be involved in the effects of cooling temperatures. We show that cooling temperatures from 35°C to 15°C decrease outward leak currents, increase input resistance, depolarize resting membrane potential (RMP), broaden AP width and increase latency of AP threshold at the NR. We further demonstrate that cooling temperatures impair regeneration of high-frequency AP trains at the NR. The effects of cooling temperatures on the intrinsic electrophysiological properties and regeneration of high-frequency AP trains at the NR can be partially reversed by BL-1249 (BL), arachidonic acid (AA), and intra-axonal protons, three thermal K2P activators, indicating the involvement of thermal K2P channels. Moreover, we show that at cooling temperatures there are interplays among thermal K2P channels, RMPs, and voltage-gated Na+ channels, which together limit regeneration of high-frequency AP trains at the NR. Our findings demonstrate a new role of thermal K2P channels in temperature-dependent conduction of high-frequency sensory signals.
Collapse
Affiliation(s)
- Hirosato Kanda
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Pharmacology, Hyogo University of Health Sciences, Kobe, Hyogo 650-8530, Japan
| | - Sotatsu Tonomura
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jianguo G Gu
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
45
|
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: 11] [Impact Index Per Article: 2.8] [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
|
46
|
Lengyel M, Enyedi P, Czirják G. Negative Influence by the Force: Mechanically Induced Hyperpolarization via K 2P Background Potassium Channels. Int J Mol Sci 2021; 22:ijms22169062. [PMID: 34445768 PMCID: PMC8396510 DOI: 10.3390/ijms22169062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 02/08/2023] Open
Abstract
The two-pore domain K2P subunits form background (leak) potassium channels, which are characterized by constitutive, although not necessarily constant activity, at all membrane potential values. Among the fifteen pore-forming K2P subunits encoded by the KCNK genes, the three members of the TREK subfamily, TREK-1, TREK-2, and TRAAK are mechanosensitive ion channels. Mechanically induced opening of these channels generally results in outward K+ current under physiological conditions, with consequent hyperpolarization and inhibition of membrane potential-dependent cellular functions. In the past decade, great advances have been made in the investigation of the molecular determinants of mechanosensation, and members of the TREK subfamily have emerged among the best-understood examples of mammalian ion channels directly influenced by the tension of the phospholipid bilayer. In parallel, the crucial contribution of mechano-gated TREK channels to the regulation of membrane potential in several cell types has been reported. In this review, we summarize the general principles underlying the mechanical activation of K2P channels, and focus on the physiological roles of mechanically induced hyperpolarization.
Collapse
|
47
|
Abstract
Migraine is a complex brain disorder explained by the interaction of genetic and environmental factors. In monogenic migraines, including familial hemiplegic migraine and migraine with aura associated with hereditary small-vessel disorders, the identified genes encode proteins expressed in neurons, astrocytes or vessels, which all increase the susceptibility to cortical spreading depression. Study of monogenic migraines showed that the neurovascular unit plays a prominent role in migraine. Genome-wide association studies have identified multiple susceptibility variants that only cause a small increase of the global migraine risk. The variants belong to several complex networks of "pro-migraine" molecular abnormalities, which are mainly neuronal or vascular. Genetics has also underscored the importance of genetic factors shared between migraine and its major co-morbidities including depression and high blood pressure. Further studies are still needed to map all of the susceptibility loci for migraine and then to understand how these genomic variants lead to migraine cell phenotypes. Thanks to the advent of new technologies such as induced pluripotent stem cells, genetic data will hopefully finally be able to lead to therapeutic progress.
Collapse
|
48
|
Huang W, Ke Y, Zhu J, Liu S, Cong J, Ye H, Guo Y, Wang K, Zhang Z, Meng W, Gao TM, Luhmann HJ, Kilb W, Chen R. TRESK channel contributes to depolarization-induced shunting inhibition and modulates epileptic seizures. Cell Rep 2021; 36:109404. [PMID: 34289346 DOI: 10.1016/j.celrep.2021.109404] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/19/2021] [Accepted: 06/23/2021] [Indexed: 11/18/2022] Open
Abstract
Glutamatergic and GABAergic synaptic transmission controls excitation and inhibition of postsynaptic neurons, whereas activity of ion channels modulates neuronal intrinsic excitability. However, it is unclear how excessive neuronal excitation affects intrinsic inhibition to regain homeostatic stability under physiological or pathophysiological conditions. Here, we report that a seizure-like sustained depolarization can induce short-term inhibition of hippocampal CA3 neurons via a mechanism of membrane shunting. This depolarization-induced shunting inhibition (DShI) mediates a non-synaptic, but neuronal intrinsic, short-term plasticity that is able to suppress action potential generation and postsynaptic responses by activated ionotropic receptors. We demonstrate that the TRESK channel significantly contributes to DShI. Disruption of DShI by genetic knockout of TRESK exacerbates the sensitivity and severity of epileptic seizures of mice, whereas overexpression of TRESK attenuates seizures. In summary, these results uncover a type of homeostatic intrinsic plasticity and its underlying mechanism. TRESK might represent a therapeutic target for antiepileptic drugs.
Collapse
Affiliation(s)
- Weiyuan Huang
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yue Ke
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianping Zhu
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuai Liu
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jin Cong
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hailin Ye
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Kewan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Center for Precision Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tian-Ming Gao
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Collaborative Innovation Center for Brain Science, Southern Medical University, Guangzhou 510515, China
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz 55120, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, Mainz 55120, Germany.
| | - Rongqing Chen
- Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
49
|
Ren K, Liu H, Guo B, Li R, Mao H, Xue Q, Yao H, Wu S, Bai Z, Wang W. Quercetin relieves D-amphetamine-induced manic-like behaviour through activating TREK-1 potassium channels in mice. Br J Pharmacol 2021; 178:3682-3695. [PMID: 33908633 DOI: 10.1111/bph.15510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Quercetin is a well-known plant flavonoid with neuroprotective properties. Earlier work suggested it may relieve psychiatric disorders, cognition deficits and memory dysfunction through anti-oxidant and/or radical scavenging mechanisms. In addition, quercetin modulated the physiological function of some ion channels. However, the detailed ionic mechanisms of the bioeffects of quercetin remain unknown. EXPERIMENTAL APPROACH Effects of quercetin on neuronal activities in the prefrontal cortex (PFC) and its ionic mechanisms were analysed by calcium imaging using mice bearing a green fluorescent protein, calmodulin, and M13 fusion protein and patch clamp in acute brain slices from C57BL/6 J mice and in HEK 293 cells. The possible ionic mechanism of action of quercetin on D-amphetamine-induced manic-like effects in mice was explored with c-fos staining and the open field behaviour test. KEY RESULTS Quercetin reduced calcium influx triggered by PFC pyramidal neuronal activity. This effect involved increasing the rheobase of neuronal firing through decreasing membrane resistance following quercetin treatment. Spadin, a blocker of TREK-1 potassium channels, also blocked the effect of quercetin on the membrane resistance and neuronal firing. Further, spadin blocked the neuroprotective effects of quercetin. The effects of quercetin on TREK-1 channels could be mimicked by GF109203X, a protein kinase C inhibitor. In vivo, injection of quercetin relieved the manic hyperlocomotion in mice, induced by D-amphetamine. This action was partly alleviated by spadin. CONCLUSION AND IMPLICATIONS TREK-1 channels are a novel target for quercetin, by inhibiting PKC. This action could contribute to both the neuroprotective and anti-manic-like effects.
Collapse
Affiliation(s)
- Keke Ren
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.,College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Haiying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Rui Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Qian Xue
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Han Yao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhantao Bai
- College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
50
|
Pavinato L, Nematian-Ardestani E, Zonta A, De Rubeis S, Buxbaum J, Mancini C, Bruselles A, Tartaglia M, Pessia M, Tucker SJ, D’Adamo MC, Brusco A. KCNK18 Biallelic Variants Associated with Intellectual Disability and Neurodevelopmental Disorders Alter TRESK Channel Activity. Int J Mol Sci 2021; 22:ijms22116064. [PMID: 34199759 PMCID: PMC8200030 DOI: 10.3390/ijms22116064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
The TWIK-related spinal cord potassium channel (TRESK) is encoded by KCNK18, and variants in this gene have previously been associated with susceptibility to familial migraine with aura (MIM #613656). A single amino acid substitution in the same protein, p.Trp101Arg, has also been associated with intellectual disability (ID), opening the possibility that variants in this gene might be involved in different disorders. Here, we report the identification of KCNK18 biallelic missense variants (p.Tyr163Asp and p.Ser252Leu) in a family characterized by three siblings affected by mild-to-moderate ID, autism spectrum disorder (ASD) and other neurodevelopment-related features. Functional characterization of the variants alone or in combination showed impaired channel activity. Interestingly, Ser252 is an important regulatory site of TRESK, suggesting that alteration of this residue could lead to additive downstream effects. The functional relevance of these mutations and the observed co-segregation in all the affected members of the family expand the clinical variability associated with altered TRESK function and provide further insight into the relationship between altered function of this ion channel and human disease.
Collapse
Affiliation(s)
- Lisa Pavinato
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
- Center for Molecular Medicine Cologne, Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany
| | - Ehsan Nematian-Ardestani
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD-2080 Msida, Malta; (E.N.-A.); (M.P.)
| | - Andrea Zonta
- Unit of Medical Genetics, “Città della Salute e della Scienza” University Hospital, 10126 Turin, Italy;
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.D.R.); (J.B.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (S.D.R.); (J.B.)
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cecilia Mancini
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy; (C.M.); (M.T.)
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy; (C.M.); (M.T.)
| | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD-2080 Msida, Malta; (E.N.-A.); (M.P.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Stephen J. Tucker
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 4BH, UK;
| | - Maria Cristina D’Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD-2080 Msida, Malta; (E.N.-A.); (M.P.)
- Correspondence: (M.C.D.); (A.B.)
| | - Alfredo Brusco
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy;
- Unit of Medical Genetics, “Città della Salute e della Scienza” University Hospital, 10126 Turin, Italy;
- Correspondence: (M.C.D.); (A.B.)
| |
Collapse
|