1
|
Fang X, Jin H, Wang J, Zhang R, Li B. Gating mechanism of the two-pore-domain potassium channel THIK1. Nat Struct Mol Biol 2025:10.1038/s41594-025-01542-4. [PMID: 40307591 DOI: 10.1038/s41594-025-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025]
Abstract
TWIK-related halothane-inhibited potassium channel (THIK1) maintains the resting membrane potential and regulates potassium efflux in microglia. It is a potential therapeutic target for neurodegenerative disorders, neuropathic pain and inflammation. However, the mechanism underlying its function remains unclear. Here we used cryo-electron microscopy to solve the structures of full-length human THIK1, revealing two inner gates and a C-type selectivity filter gate, distinct from other two-pore-domain potassium channels. One inner gate, formed by a short helix in the distal C terminus, introduces a unique gating mechanism involving the distal cytoplasmic domain. The other, beneath the selectivity filter, is constricted by Y273 in the M4 helix, dividing the cavity. In addition, the selectivity filter gate is modulated by polyunsaturated fatty acids. These structural insights into THIK1 gating, through the distal C-terminal helices, hydrophilic residues and selectivity filter, advance our understanding of THIK1's role in microglial homeostasis and neuropathologies.
Collapse
Affiliation(s)
- Xiangyun Fang
- Department of Anesthesiology, Fudan University, Shanghai, China
- Institute for Translational Brain Research, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Haichao Jin
- School of Science, China Pharmaceutical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jin Wang
- School of Science, China Pharmaceutical University, Nanjing, China.
| | - Ran Zhang
- Department of Anesthesiology, Fudan University, Shanghai, China.
- Institute for Translational Brain Research, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Baobin Li
- Department of Anesthesiology, Fudan University, Shanghai, China.
- Institute for Translational Brain Research, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
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
|
Zhuang W, Mun SY, Park WS. Direct effects of antipsychotics on potassium channels. Biochem Biophys Res Commun 2025; 749:151344. [PMID: 39842331 DOI: 10.1016/j.bbrc.2025.151344] [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: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) and are severe psychiatric conditions that contribute to disability and increased healthcare costs globally. Although first-, second-, and third-generation antipsychotics are available for treating BD and SCZ, most have various side effects unrelated to their unique functions. Many antipsychotics affect K+ channels (Kv, KCa, Kir, K2P, and other channels), which change the functions of various organs. This review summarizes the biological actions of antipsychotics, including off-target side effects involving K+ channels.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
4
|
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
|
5
|
Krygier M, Ziętkiewicz S, Talaśka-Liczbik W, Chylińska M, Walczak A, Kostrzewa G, Płoski R, Mazurkiewicz-Bełdzińska M. The epilepsy phenotype of KCNK4-related neurodevelopmental disease. Seizure 2024; 121:114-122. [PMID: 39146707 DOI: 10.1016/j.seizure.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
INTRODUCTION Potassium ion channels play a crucial role in maintaining cellular electrical stability and are implicated in various epilepsies. Heterozygous pathogenic variants in KCNK4 cause a recognizable neurodevelopmental syndrome with facial dysmorphism, hypertrichosis, epilepsy, intellectual disability (ID), and gingival overgrowth (FHEIG). To date, no more than nine patients with FHEIG have been described worldwide and still little is known about epileptic phenotype in KCNK4-related disease. METHODS We identified a novel de novo p.(Gly139Arg) variant in KCNK4 in a patient with drug-resistant nocturnal seizures, mild ID, and dysmorphic features. In silico analyses of the variant strongly suggest a gain-of-function effect. We conducted a retrospective review of previously published cases, focusing on the epileptic features and response to various treatments. RESULTS To date, epilepsy has been reported in 8/10 patients with KCNK4-related disease. The mean age of seizure onset was 1.8 years, and the most common seizure type was focal to bilateral tonic-clonic (5/8). Sodium channel blockers and valproate were effective in the majority of patients, but in 3/8 the epilepsy was drug-resistant. Our patient showed improved seizure control after treatment with the carbonic anhydrase inhibitor sulthiame. Interestingly, the patient showed features of peripheral nerve hyperexcitability syndrome, a phenomenon not previously described in potassium channelopathies caused by increased K+ conductance. CONCLUSION Gain-of-function variants in KCNK4 cause a spectrum of epilepsies, ranging from benign isolated epilepsy to epileptic encephalopathy, with focal to bilateral tonic-clonic seizures being the most commonly observed. Importantly, a subgroup of patients present with a mild extra-neurological phenotype without characteristic facial dysmorphism or generalized hypertrichosis. This report expands the phenotypic spectrum of KNCK4-associated disease and provides new insights into the clinical heterogeneity of this rare neurodevelopmental syndrome.
Collapse
Affiliation(s)
- Magdalena Krygier
- Department of Developmental Neurology, Medical University of Gdansk, Gdansk, Poland.
| | - Szymon Ziętkiewicz
- Laboratory of Protein Biochemistry, Intercollegiate Faculty of Biotechnology, University of Gdansk, Gdansk, Poland
| | | | | | - Anna Walczak
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Grażyna Kostrzewa
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
6
|
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
|
7
|
Türkaydin B, Schewe M, Riel EB, Schulz F, Biedermann J, Baukrowitz T, Sun H. Atomistic mechanism of coupling between cytosolic sensor domain and selectivity filter in TREK K2P channels. Nat Commun 2024; 15:4628. [PMID: 38821927 PMCID: PMC11143257 DOI: 10.1038/s41467-024-48823-y] [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: 10/13/2023] [Accepted: 05/15/2024] [Indexed: 06/02/2024] Open
Abstract
The two-pore domain potassium (K2P) channels TREK-1 and TREK-2 link neuronal excitability to a variety of stimuli including mechanical force, lipids, temperature and phosphorylation. This regulation involves the C-terminus as a polymodal stimulus sensor and the selectivity filter (SF) as channel gate. Using crystallographic up- and down-state structures of TREK-2 as a template for full atomistic molecular dynamics (MD) simulations, we reveal that the SF in down-state undergoes inactivation via conformational changes, while the up-state structure maintains a stable and conductive SF. This suggests an atomistic mechanism for the low channel activity previously assigned to the down state, but not evident from the crystal structure. Furthermore, experimentally by using (de-)phosphorylation mimics and chemically attaching lipid tethers to the proximal C-terminus (pCt), we confirm the hypothesis that moving the pCt towards the membrane induces the up-state. Based on MD simulations, we propose two gating pathways by which movement of the pCt controls the stability (i.e., conductivity) of the filter gate. Together, these findings provide atomistic insights into the SF gating mechanism and the physiological regulation of TREK channels by phosphorylation.
Collapse
Affiliation(s)
- Berke Türkaydin
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Insitute of Chemistry, Technical University of Berlin, Berlin, Germany
| | - Marcus Schewe
- Institute of Physiology, Kiel University, Kiel, Germany.
| | - Elena Barbara Riel
- Institute of Physiology, Kiel University, Kiel, Germany
- Department of Anesthesiology, Weill Cornell Medical College, New York, USA
| | | | - Johann Biedermann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.
- Insitute of Chemistry, Technical University of Berlin, Berlin, Germany.
| |
Collapse
|
8
|
Mӓnnikkӧ R, Kullmann DM. Structure-function and pharmacologic aspects of ion channels relevant to neurologic channelopathies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:1-23. [PMID: 39174242 DOI: 10.1016/b978-0-323-90820-7.00009-4] [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] [Indexed: 08/24/2024]
Abstract
Ion channels are membrane proteins that allow the passage of ions across the membrane. They characteristically contain a pore where the selectivity of certain ion species is determined and gates that open and close the pore are found. The pore is often connected to additional domains or subunits that regulate its function. Channels are grouped into families based on their selectivity for specific ions and the stimuli that control channel opening and closing, such as voltage or ligands. Ion channels are fundamental to the electrical properties of excitable tissues. Dysfunction of channels can lead to abnormal electrical signaling of neurons and muscle cells, accompanied by clinical manifestations, known as channelopathies. Many naturally occurring toxins target ion channels and affect excitable cells where the channels are expressed. Furthermore, ion channels, as membrane proteins and key regulators of a number of physiologic functions, are an important target for drugs in clinical use. In this chapter, we give a general overview of the classification, genetics and structure-function features of the main ion channel families, and address some pharmacologic aspects relevant to neurologic channelopathies.
Collapse
Affiliation(s)
- Roope Mӓnnikkӧ
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
9
|
Zhu Y, Peng BJ, Kumar S, Stover L, Chang JY, Lyu J, Zhang T, Schrecke S, Azizov D, Russell DH, Fang L, Laganowsky A. Polyamine detergents tailored for native mass spectrometry studies of membrane proteins. Nat Commun 2023; 14:5676. [PMID: 37709761 PMCID: PMC10502129 DOI: 10.1038/s41467-023-41429-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Native mass spectrometry (MS) is a powerful technique for interrogating membrane protein complexes and their interactions with other molecules. A key aspect of the technique is the ability to preserve native-like structures and noncovalent interactions, which can be challenging depending on the choice of detergent. Different strategies have been employed to reduce charge on protein complexes to minimize activation and preserve non-covalent interactions. Here, we report the synthesis of a class of polyamine detergents tailored for native MS studies of membrane proteins. These detergents, a series of spermine covalently attached to various alkyl tails, are exceptional charge-reducing molecules, exhibiting a ten-fold enhanced potency over spermine. Addition of polyamine detergents to proteins solubilized in maltoside detergents results in improved, charge-reduced native mass spectra and reduced dissociation of subunits. Polyamine detergents open new opportunities to investigate membrane proteins in different detergent environments that have thwarted previous native MS studies.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Bo-Ji Peng
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Smriti Kumar
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Lauren Stover
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Jing-Yuan Chang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Samantha Schrecke
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Djavdat Azizov
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - David H Russell
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Lei Fang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA.
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
10
|
Arévalo B, Bedoya M, Kiper AK, Vergara F, Ramírez D, Mazola Y, Bustos D, Zúñiga R, Cikutovic R, Cayo A, Rinné S, Ramirez-Apan MT, Sepúlveda FV, Cerda O, López-Collazo E, Decher N, Zúñiga L, Gutierrez M, González W. Selective TASK-1 Inhibitor with a Defined Structure–Activity Relationship Reduces Cancer Cell Proliferation and Viability. J Med Chem 2022; 65:15014-15027. [DOI: 10.1021/acs.jmedchem.1c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bárbara Arévalo
- Centro de Estudios en Alimentos Procesados−CEAP, Conicyt, Programa Regional R19A10001, Gore Maule, 3460000 Talca, Chile
| | - Mauricio Bedoya
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, 3460000 Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, 3480094 Talca, Chile
| | - Aytug K. Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
| | - Fernando Vergara
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| | - David Ramírez
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, 4030000 Concepción, Chile
| | - Yuliet Mazola
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, 3460000 Talca, Chile
- Laboratorio de Bioinformática y Química Computacional (LBQC), Escuela de Química y Farmacia, Facultad de Medicina, Universidad Católica del Maule, 3460000 Talca, Chile
| | - Rafael Zúñiga
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
- Instituto de Investigación Interdisciplinaria, Vicerrectoría Académica, Universidad de Talca, 3460000 Talca, Chile
| | - Rocio Cikutovic
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Angel Cayo
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
| | - M. Teresa Ramirez-Apan
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510, Coyoacán, 04510 México, DF, México
| | - Francisco V. Sepúlveda
- Centro de Estudios Científicos (CECs), 5110466 Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, 5110466 Valdivia, Chile
| | - Oscar Cerda
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, 8380453 Santiago, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Facultad de Medicina, Universidad de Chile, 8380453 Santiago, Chile
| | - Eduardo López-Collazo
- The Innate Immune Response Group and Tumor Immunology Laboratory, IdiPAZ, La Paz University Hospital, 8046 Madrid, Spain
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 1-2, 35037 Marburg, Germany
- Marburg Center for Mind, Brain and Behavior−MCMBB, Philipps-University Marburg, 35037 Marburg, Germany
| | - Leandro Zúñiga
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Casilla, 3460000 Talca, Chile
| | - Margarita Gutierrez
- Laboratorio de Síntesis y Actividad Biológica, Instituto de Química de Recursos Naturales, Universidad de Talca, 1 poniente No. 1141, 3460000 Talca, Chile
| | - Wendy González
- Centro de Bioinformática, Simulación y Modelado (CBSM), Facultad de Ingeniería, Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, 1 Poniente No. 1141, 3460000 Talca, Chile
| |
Collapse
|
11
|
Burata OE, Yeh TJ, Macdonald CB, Stockbridge RB. Still rocking in the structural era: A molecular overview of the small multidrug resistance (SMR) transporter family. J Biol Chem 2022; 298:102482. [PMID: 36100040 PMCID: PMC9574504 DOI: 10.1016/j.jbc.2022.102482] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/24/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
Abstract
The small multidrug resistance (SMR) family is composed of widespread microbial membrane proteins that fulfill different transport functions. Four functional SMR subtypes have been identified, which variously transport the small, charged metabolite guanidinium, bulky hydrophobic drugs and antiseptics, polyamines, and glycolipids across the membrane bilayer. The transporters possess a minimalist architecture, with ∼100-residue subunits that require assembly into homodimers or heterodimers for transport. In part because of their simple construction, the SMRs are a tractable system for biochemical and biophysical analysis. Studies of SMR transporters over the last 25 years have yielded deep insights for diverse fields, including membrane protein topology and evolution, mechanisms of membrane transport, and bacterial multidrug resistance. Here, we review recent advances in understanding the structures and functions of SMR transporters. New molecular structures of SMRs representing two of the four functional subtypes reveal the conserved structural features that have permitted the emergence of disparate substrate transport functions in the SMR family and illuminate structural similarities with a distantly related membrane transporter family, SLC35/DMT.
Collapse
Affiliation(s)
- Olive E Burata
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Trevor Justin Yeh
- Program in Biophysics, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Randy B Stockbridge
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, USA; Program in Biophysics, University of Michigan, Ann Arbor, Michigan, USA; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
12
|
Turney TS, Li V, Brohawn SG. Structural Basis for pH-gating of the K + channel TWIK1 at the selectivity filter. Nat Commun 2022; 13:3232. [PMID: 35680900 PMCID: PMC9184524 DOI: 10.1038/s41467-022-30853-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/20/2022] [Indexed: 11/11/2022] Open
Abstract
TWIK1 (K2P1.1, KCNK1) is a widely expressed pH-gated two-pore domain K+ channel (K2P) that contributes to cardiac rhythm generation and insulin release from pancreatic beta cells. TWIK1 displays unique properties among K2Ps including low basal activity and inhibition by extracellular protons through incompletely understood mechanisms. Here, we present cryo-EM structures of TWIK1 in lipid nanodiscs at high and low pH that reveal a previously undescribed gating mechanism at the K+ selectivity filter. At high pH, TWIK1 adopts an open conformation. At low pH, protonation of an extracellular histidine results in a cascade of conformational changes that close the channel by sealing the top of the selectivity filter, displacing the helical cap to block extracellular ion access pathways, and opening gaps for lipid block of the intracellular cavity. These data provide a mechanistic understanding for extracellular pH-gating of TWIK1 and illustrate how diverse mechanisms have evolved to gate the selectivity filter of K+ channels.
Collapse
Affiliation(s)
- Toby S Turney
- Biophysics Graduate Program, University of California Berkeley, Berkeley, CA, 94720, USA
- 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 Biosciences (QB3), University of California Berkeley, Berkeley, CA, 94720, USA
| | - Vivian 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 Biosciences (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 Biosciences (QB3), University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
13
|
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
|
14
|
Zhang Q, Fu J, Zhang S, Guo P, Liu S, Shen J, Guo J, Yang H. 'C-type' closed state and gating mechanisms of K2P channels revealed by conformational changes of the TREK-1 channel. J Mol Cell Biol 2022; 14:6504012. [PMID: 35022758 PMCID: PMC9021975 DOI: 10.1093/jmcb/mjac002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 01/10/2023] Open
Abstract
Two-pore domain potassium (K2P) channels gate primarily within the selectivity filter, termed ‘C-type’ gating. Due to the lack of structural insights into the nonconductive (closed) state, ‘C-type’ gating mechanisms remain elusive. Here, molecular dynamics (MD) simulations on TREK-1, a K2P channel, revealed that M4 helix movements induce filter closing in a novel ‘deeper-down’ structure that represents a ‘C-type’ closed state. The ‘down’ structure does not represent the closed state as previously proposed and instead acts as an intermediate state in gating. The study identified the allosteric ‘seesaw’ mechanism of M4 helix movements in modulating filter closing. Finally, guided by this recognition of K2P gating mechanisms, MD simulations revealed that gain-of-function mutations and small-molecule activators activate TREK-1 by perturbing state transitions from open to closed states. Together, we reveal a ‘C-type’ closed state and provide mechanical insights into gating procedures and allosteric regulations for K2P channels.
Collapse
Affiliation(s)
- Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Fu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shaoying Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peipei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Shijie Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juwen Shen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiangtao Guo
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
15
|
García-Fernández MD, Chatelain FC, Nury H, Moroni A, Moreau CJ. Distinct classes of potassium channels fused to GPCRs as electrical signaling biosensors. CELL REPORTS METHODS 2021; 1:None. [PMID: 34977850 PMCID: PMC8688152 DOI: 10.1016/j.crmeth.2021.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/05/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022]
Abstract
Ligand-gated ion channels (LGICs) are natural biosensors generating electrical signals in response to the binding of specific ligands. Creating de novo LGICs for biosensing applications is technically challenging. We have previously designed modified LGICs by linking G protein-coupled receptors (GPCRs) to the Kir6.2 channel. In this article, we extrapolate these design concepts to other channels with different structures and oligomeric states, namely a tetrameric viral Kcv channel and the dimeric mouse TREK-1 channel. After precise engineering of the linker regions, the two ion channels were successfully regulated by a GPCR fused to their N-terminal domain. Two-electrode voltage-clamp recordings showed that Kcv and mTREK-1 fusions were inhibited and activated by GPCR agonists, respectively, and antagonists abolished both effects. Thus, dissimilar ion channels can be allosterically regulated through their N-terminal domains, suggesting that this is a generalizable approach for ion channel engineering.
Collapse
Affiliation(s)
| | - Franck C. Chatelain
- Université Côte d’Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles, 06650 Valbonne, France
| | - Hugues Nury
- Université Grenoble Alpes, CNRS, CEA, IBS, 71, av. Martyrs, CS10090, 38044 Grenoble Cedex9, France
| | - Anna Moroni
- University of Milan, Department of Biosciences, Via Celoria 26, 20133 Milano, Italy
| | - Christophe J. Moreau
- Université Grenoble Alpes, CNRS, CEA, IBS, 71, av. Martyrs, CS10090, 38044 Grenoble Cedex9, France
| |
Collapse
|
16
|
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: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/10/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
TRAAK is a mechanosensitive two-pore domain K+ (K2P) channel localized to nodes of Ranvier in myelinated neurons. TRAAK deletion in mice results in mechanical and thermal allodynia, and gain-of-function mutations cause the human neurodevelopmental disorder FHEIG. TRAAK displays basal and stimulus-gated activities typical of K2Ps, but the mechanistic and structural differences between these modes are unknown. Here, we demonstrate that basal and mechanically gated openings are distinguished by their conductance, kinetics, and structure. Basal openings are low conductance, short duration, and due to a conductive channel conformation with the interior cavity exposed to the surrounding membrane. Mechanically gated openings are high conductance, long duration, and due to a channel conformation in which the interior cavity is sealed to the surrounding membrane. Our results explain how dual modes of activity are produced by a single ion channel and provide a basis for the development of state-selective pharmacology with the potential to treat disease.
Collapse
Affiliation(s)
- Robert A Rietmeijer
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Program, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Ben Sorum
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Baobin Li
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA
| | - Stephen G Brohawn
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biology (QB3), University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
17
|
Tahir S, Bourquard T, Musnier A, Jullian Y, Corde Y, Omahdi Z, Mathias L, Reiter E, Crépieux P, Bruneau G, Poupon A. Accurate determination of epitope for antibodies with unknown 3D structures. MAbs 2021; 13:1961349. [PMID: 34432559 PMCID: PMC8405158 DOI: 10.1080/19420862.2021.1961349] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MAbTope is a docking-based method for the determination of epitopes. It has been used to successfully determine the epitopes of antibodies with known 3D structures. However, during the antibody discovery process, this structural information is rarely available. Although we already have evidence that homology models of antibodies could be used instead of their 3D structure, the choice of the template, the methodology for homology modeling and the resulting performance still have to be clarified. Here, we show that MAbTope has the same performance when working with homology models of the antibodies as compared to crystallographic structures. Moreover, we show that even low-quality models can be used. We applied MAbTope to determine the epitope of dupilumab, an anti- interleukin 4 receptor alpha subunit therapeutic antibody of unknown 3D structure, that we validated experimentally. Finally, we show how the MAbTope-determined epitopes for a series of antibodies targeting the same protein can be used to predict competitions, and demonstrate the accuracy with an experimentally validated example. 3D: three-dimensionalRMSD: root mean square deviationCDR: complementary-determining regionCPU: central processing unitsVH: heavy chain variable regionVL: light chain variable regionscFv: single-chain variable fragmentsVHH: single-chain antibody variable regionIL4Rα: Interleukin 4 receptor alpha chainSPR: surface plasmon resonancePDB: protein data bankHEK293: Human embryonic kidney 293 cellsEDTA: Ethylenediaminetetraacetic acidFBS: Fetal bovine serumANOVA: Analysis of varianceEGFR: Epidermal growth factor receptorPE: PhycoerythrinAPC: AllophycocyaninFSC: forward scatterSSC: side scatterWT: wild type Keywords: MAbTope, Epitope Mapping, Molecular docking, Antibody modeling, Antibody-antigen docking
Collapse
Affiliation(s)
- Shifa Tahir
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France
| | - Thomas Bourquard
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais
| | - Astrid Musnier
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais
| | - Yann Jullian
- MAbSilico SAS, 1 Impasse Du Palais.,CaSciModOT, UFR De Sciences Et Techniques, Université De Tours
| | | | | | | | - Eric Reiter
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | - Pascale Crépieux
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | | | - Anne Poupon
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| |
Collapse
|
18
|
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
|
19
|
Domene C, Ocello R, Masetti M, Furini S. Ion Conduction Mechanism as a Fingerprint of Potassium Channels. J Am Chem Soc 2021; 143:12181-12193. [PMID: 34323472 DOI: 10.1021/jacs.1c04802] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
K+-channels are membrane proteins that regulate the selective conduction of potassium ions across cell membranes. Although the atomic mechanisms of K+ permeation have been extensively investigated, previous work focused on characterizing the selectivity and occupancy of the binding sites, the role of water molecules in the conduction process, or the identification of the minimum energy pathways enabling permeation. Here, we exploit molecular dynamics simulations and the analytical power of Markov state models to perform a comparative study of ion conduction in three distinct channel models. Significant differences emerged in terms of permeation mechanisms and binding site occupancy by potassium ions and/or water molecules from 100 μs cumulative trajectories. We found that, at odds with the current paradigm, each system displays a characteristic permeation mechanism, and thus, there is not a unique way by which potassium ions move through K+-channels. The high functional diversity of K+-channels can be attributed in part to the differences in conduction features that have emerged from this work. This study provides crucial information and further inspiration for wet-lab chemists designing new synthetic strategies to produce versatile artificial ion channels that emulate membrane transport for their applications in diagnosis, sensors, the next generation of water treatment technologies, etc., as the ability of synthetic channels to transport molecular ions across a bilayer in a controlled way is usually governed through the choice of metal ions, their oxidation states, or their coordination geometries.
Collapse
Affiliation(s)
- Carmen Domene
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, U.K.,Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, U.K
| | - Riccardo Ocello
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Università di Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| |
Collapse
|
20
|
Cunningham KP, Clapp LH, Mathie A, Veale EL. The Prostacyclin Analogue, Treprostinil, Used in the Treatment of Pulmonary Arterial Hypertension, is a Potent Antagonist of TREK-1 and TREK-2 Potassium Channels. Front Pharmacol 2021; 12:705421. [PMID: 34267666 PMCID: PMC8276018 DOI: 10.3389/fphar.2021.705421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an aggressive vascular remodeling disease that carries a high morbidity and mortality rate. Treprostinil (Remodulin) is a stable prostacyclin analogue with potent vasodilatory and anti-proliferative activity, approved by the FDA and WHO as a treatment for PAH. A limitation of this therapy is the severe subcutaneous site pain and other forms of pain experienced by some patients, which can lead to significant non-compliance. TWIK-related potassium channels (TREK-1 and TREK-2) are highly expressed in sensory neurons, where they play a role in regulating sensory neuron excitability. Downregulation, inhibition or mutation of these channels leads to enhanced pain sensitivity. Using whole-cell patch-clamp electrophysiological recordings, we show, for the first time, that treprostinil is a potent antagonist of human TREK-1 and TREK-2 channels but not of TASK-1 channels. An increase in TASK-1 channel current was observed with prolonged incubation, consistent with its therapeutic role in PAH. To investigate treprostinil-induced inhibition of TREK, site-directed mutagenesis of a number of amino acids, identified as important for the action of other regulatory compounds, was carried out. We found that a gain of function mutation of TREK-1 (Y284A) attenuated treprostinil inhibition, while a selective activator of TREK channels, BL-1249, overcame the inhibitory effect of treprostinil. Our data suggests that subcutaneous site pain experienced during treprostinil therapy may result from inhibition of TREK channels near the injection site and that pre-activation of these channels prior to treatment has the potential to alleviate this nociceptive activity.
Collapse
Affiliation(s)
- Kevin P Cunningham
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom.,Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Lucie H Clapp
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom.,School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, United Kingdom
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent and University of Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
21
|
Abstract
Monoclonal antibodies combine specificity and high affinity binding with excellent pharmacokinetic properties and are rapidly being developed for a wide range of drug targets including clinically important potassium ion channels. Nonetheless, while therapeutic antibodies come with great promise, K+ channels represent particularly difficult targets for biologics development for a variety of reasons that include their dynamic structures and relatively small extracellular loops, their high degree of sequence conservation (leading to immune tolerance), and their generally low-level expression in vivo. The process is made all the more difficult when large numbers of antibody candidates must be screened for a given target, or when lead candidates fail to cross-react with orthologous channels in animal disease models due to their highly selective binding properties. While the number of antibodies targeting potassium channels in preclinical or clinical development is still modest, significant advances in the areas of protein expression and antibody screening are converging to open the field to an avalanche of new drugs. Here, the opportunities and constraints associated with the discovery of antibodies against K+ channels are discussed, with an emphasis on novel technologies that are opening the field to exciting new possibilities for biologics development.
Collapse
|
22
|
Natale AM, Deal PE, Minor DL. Structural Insights into the Mechanisms and Pharmacology of K 2P Potassium Channels. J Mol Biol 2021; 433:166995. [PMID: 33887333 PMCID: PMC8436263 DOI: 10.1016/j.jmb.2021.166995] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/10/2023]
Abstract
Leak currents, defined as voltage and time independent flows of ions across cell membranes, are central to cellular electrical excitability control. The K2P (KCNK) potassium channel class comprises an ion channel family that produces potassium leak currents that oppose excitation and stabilize the resting membrane potential in cells in the brain, cardiovascular system, immune system, and sensory organs. Due to their widespread tissue distribution, K2Ps contribute to many physiological and pathophysiological processes including anesthesia, pain, arrythmias, ischemia, hypertension, migraine, intraocular pressure regulation, and lung injury responses. Structural studies of six homomeric K2Ps have established the basic architecture of this channel family, revealed key moving parts involved in K2P function, uncovered the importance of asymmetric pinching and dilation motions in the K2P selectivity filter (SF) C-type gate, and defined two K2P structural classes based on the absence or presence of an intracellular gate. Further, a series of structures characterizing K2P:modulator interactions have revealed a striking polysite pharmacology housed within a relatively modestly sized (~70 kDa) channel. Binding sites for small molecules or lipids that control channel function are found at every layer of the channel structure, starting from its extracellular side through the portion that interacts with the membrane bilayer inner leaflet. This framework provides the basis for understanding how gating cues sensed by different channel parts control function and how small molecules and lipids modulate K2P activity. Such knowledge should catalyze development of new K2P modulators to probe function and treat a wide range of disorders.
Collapse
Affiliation(s)
- Andrew M Natale
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Parker E Deal
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience University of California, San Francisco, CA 94158, USA; Molecular Biophysics and Integrated Bio-imaging Division Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
23
|
Lee H, Lolicato M, Arrigoni C, Minor DL. Production of K 2P2.1 (TREK-1) for structural studies. Methods Enzymol 2021; 653:151-188. [PMID: 34099170 DOI: 10.1016/bs.mie.2021.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
K2P (KCNK) potassium channels form 'background' or 'leak' currents that are important for controlling cell excitability in the brain, cardiovascular system, and somatosensory neurons. K2P2.1 (TREK-1) is one of the founding members of this family and one of the first well-characterized polymodal ion channels capable of responding to a variety of physical and chemical gating cues. Of the six K2P subfamilies, the thermo-and mechano-sensitive TREK subfamily comprising K2P2.1 (TREK-1), K2P4.1 (TRAAK), and K2P10.1 (TREK-2) is the first to have structures determined for each subfamily member. These structural studies have revealed key architectural features that provide a framework for understanding how gating cues sensed by different channel elements converge on the K2P selectivity filter C-type gate. TREK family structural studies have also revealed numerous sites where small molecules or lipids bind and affect channel function. This rich structural landscape provides the framework for probing K2P function and for the development of new K2P-directed agents. Such molecules may be useful for affecting processes where TREK channels are important such as anesthesia, pain, arrythmia, ischemia, migraine, intraocular pressure, and lung injury. Production of high quality protein samples is key to addressing new questions about K2P function and pharmacology. Here, we present methods for producing pure K2P2.1 (TREK-1) suitable for advancing towards these goals through structural and biochemical studies.
Collapse
Affiliation(s)
- Haerim Lee
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Cristina Arrigoni
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States; Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA, United States; California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA, United States; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, United States; Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
24
|
Khoubza L, Chatelain FC, Feliciangeli S, Lesage F, Bichet D. Physiological roles of heteromerization: focus on the two-pore domain potassium channels. J Physiol 2021; 599:1041-1055. [PMID: 33347640 DOI: 10.1113/jp279870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
Potassium channels form the largest family of ion channels with more than 80 members involved in cell excitability and signalling. Most of them exist as homomeric channels, whereas specific conditions are required to obtain heteromeric channels. It is well established that heteromerization of voltage-gated and inward rectifier potassium channels affects their function, increasing the diversity of the native potassium currents. For potassium channels with two pore domains (K2P ), homomerization has long been considered the rule, their polymodal regulation by a wide diversity of physical and chemical stimuli being responsible for the adaptation of the leak potassium currents to cellular needs. This view has recently evolved with the accumulation of evidence of heteromerization between different K2P subunits. Several functional intragroup and intergroup heteromers have recently been identified, which contribute to the functional heterogeneity of this family. K2P heteromerization is involved in the modulation of channel expression and trafficking, promoting functional and signalling diversity. As illustrated in the Abstract Figure, heteromerization of TREK1 and TRAAK provides the cell with more possibilities of regulation. It is becoming increasingly evident that K2P heteromers contribute to important physiological functions including neuronal and cardiac excitability. Since heteromerization also affects the pharmacology of K2P channels, this understanding helps to establish K2P heteromers as new therapeutic targets for physiopathological conditions.
Collapse
Affiliation(s)
- Lamyaa Khoubza
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| | - Franck C Chatelain
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| | - Sylvain Feliciangeli
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France.,Inserm, 101 rue de Tolbiac, 75013, Paris, France
| | - Florian Lesage
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France.,Inserm, 101 rue de Tolbiac, 75013, Paris, France
| | - Delphine Bichet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| |
Collapse
|
25
|
Abstract
Potassium channels are present in every living cell and essential to setting up a stable, non-zero transmembrane electrostatic potential which manifests the off-equilibrium livelihood of the cell. They are involved in other cellular activities and regulation, such as the controlled release of hormones, the activation of T-cells for immune response, the firing of action potential in muscle cells and neurons, etc. Pharmacological reagents targeting potassium channels are important for treating various human diseases linked to dysfunction of the channels. High-resolution structures of these channels are very useful tools for delineating the detailed chemical basis underlying channel functions and for structure-based design and optimization of their pharmacological and pharmaceutical agents. Structural studies of potassium channels have revolutionized biophysical understandings of key concepts in the field - ion selectivity, conduction, channel gating, and modulation, making them multi-modality targets of pharmacological regulation. In this chapter, I will select a few high-resolution structures to illustrate key structural insights, proposed allostery behind channel functions, disagreements still open to debate, and channel-lipid interactions and co-evolution. The known structural consensus allows the inference of conserved molecular mechanisms shared among subfamilies of K+ channels and makes it possible to develop channel-specific pharmaceutical agents.
Collapse
Affiliation(s)
- Qiu-Xing Jiang
- Laboratory of Molecular Physiology and Biophysics and the Cryo-EM Center, Hauptmann-Woodward Medical Research Institute, Buffalo, NY, USA.
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA.
- Departments of Materials Design and Invention and Physiology and Biophysics, University of Buffalo (SUNY), Buffalo, NY, USA.
| |
Collapse
|
26
|
Ocello R, Furini S, Lugli F, Recanatini M, Domene C, Masetti M. Conduction and Gating Properties of the TRAAK Channel from Molecular Dynamics Simulations with Different Force Fields. J Chem Inf Model 2020; 60:6532-6543. [PMID: 33295174 PMCID: PMC8016162 DOI: 10.1021/acs.jcim.0c01179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 12/20/2022]
Abstract
In recent years, the K2P family of potassium channels has been the subject of intense research activity. Owing to the complex function and regulation of this family of ion channels, it is common practice to complement experimental findings with the atomistic description provided by computational approaches such as molecular dynamics (MD) simulations, especially, in light of the unprecedented timescales accessible at present. However, despite recent substantial improvements, the accuracy of MD simulations is still undermined by the intrinsic limitations of force fields. Here, we systematically assessed the performance of the most popular force fields employed to study ion channels at timescales that are orders of magnitude greater than the ones accessible when these energy functions were first developed. Using 32 μs of trajectories, we investigated the dynamics of a member of the K2P ion channel family, the TRAAK channel, using two established force fields in simulations of biological systems: AMBER and CHARMM. We found that while results are comparable on the nanosecond timescales, significant inconsistencies arise at microsecond timescales.
Collapse
Affiliation(s)
- Riccardo Ocello
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum−Università di Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Simone Furini
- Department
of Medical Biotechnologies, University of
Siena, 53100 Siena, Italy
| | - Francesca Lugli
- Department
of Chemistry “G. Ciamician”, Alma Mater Studiorum—Università di Bologna, via Selmi 2, 40126 Bologna, Italy
| | - Maurizio Recanatini
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum−Università di Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Carmen Domene
- Department
of Chemistry, University of Bath, Claverton Down, BA2 7AY Bath, U.K.
- Department
of Chemistry, University of Oxford, Mansfield Road, OX1 3TA Oxford, U.K.
| | - Matteo Masetti
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum−Università di Bologna, via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
27
|
Discoveries in structure and physiology of mechanically activated ion channels. Nature 2020; 587:567-576. [PMID: 33239794 DOI: 10.1038/s41586-020-2933-1] [Citation(s) in RCA: 343] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/19/2020] [Indexed: 01/24/2023]
Abstract
The ability to sense physical forces is conserved across all organisms. Cells convert mechanical stimuli into electrical or chemical signals via mechanically activated ion channels. In recent years, the identification of new families of mechanosensitive ion channels-such as PIEZO and OSCA/TMEM63 channels-along with surprising insights into well-studied mechanosensitive channels have driven further developments in the mechanotransduction field. Several well-characterized mechanosensory roles such as touch, blood-pressure sensing and hearing are now linked with primary mechanotransducers. Unanticipated roles of mechanical force sensing continue to be uncovered. Furthermore, high-resolution structures representative of nearly every family of mechanically activated channel described so far have underscored their diversity while advancing our understanding of the biophysical mechanisms of pressure sensing. Here we summarize recent discoveries in the physiology and structures of known mechanically activated ion channel families and discuss their implications for understanding the mechanisms of mechanical force sensing.
Collapse
|
28
|
Lolicato M, Natale AM, Abderemane-Ali F, Crottès D, Capponi S, Duman R, Wagner A, Rosenberg JM, Grabe M, Minor DL. K 2P channel C-type gating involves asymmetric selectivity filter order-disorder transitions. SCIENCE ADVANCES 2020; 6:6/44/eabc9174. [PMID: 33127683 PMCID: PMC7608817 DOI: 10.1126/sciadv.abc9174] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/10/2020] [Indexed: 05/05/2023]
Abstract
K2P potassium channels regulate cellular excitability using their selectivity filter (C-type) gate. C-type gating mechanisms, best characterized in homotetrameric potassium channels, remain controversial and are attributed to selectivity filter pinching, dilation, or subtle structural changes. The extent to which such mechanisms control C-type gating of innately heterodimeric K2Ps is unknown. Here, combining K2P2.1 (TREK-1) x-ray crystallography in different potassium concentrations, potassium anomalous scattering, molecular dynamics, and electrophysiology, we uncover unprecedented, asymmetric, potassium-dependent conformational changes that underlie K2P C-type gating. These asymmetric order-disorder transitions, enabled by the K2P heterodimeric architecture, encompass pinching and dilation, disrupt the S1 and S2 ion binding sites, require the uniquely long K2P SF2-M4 loop and conserved "M3 glutamate network," and are suppressed by the K2P C-type gate activator ML335. These findings demonstrate that two distinct C-type gating mechanisms can operate in one channel and underscore the SF2-M4 loop as a target for K2P channel modulator development.
Collapse
Affiliation(s)
- Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA
| | - Andrew M Natale
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA
| | - Fayal Abderemane-Ali
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA
| | - David Crottès
- Department of Physiology, University of California, San Francisco, CA 93858-2330, USA
| | - Sara Capponi
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA
| | - Ramona Duman
- Science Division, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - Armin Wagner
- Science Division, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| | - John M Rosenberg
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Michael Grabe
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 93858-2330, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, CA 93858-2330, USA.
- Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, CA 93858-2330, USA
- California Institute for Quantitative Biomedical Research, University of California, San Francisco, CA 93858-2330, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA, 93858-2330, USA
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720 USA
| |
Collapse
|
29
|
Selective regulation of human TRAAK channels by biologically active phospholipids. Nat Chem Biol 2020; 17:89-95. [PMID: 32989299 PMCID: PMC7746637 DOI: 10.1038/s41589-020-00659-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/21/2020] [Indexed: 11/22/2022]
Abstract
TRAAK is an ion channel from the two-pore domain potassium (K2P) channel family with roles in maintaining the resting membrane potential and fast action potential conduction. Regulated by a wide range of physical and chemical stimuli, the affinity and selectivity of K2P4.1 towards lipids remains poorly understood. Here we show the two isoforms of K2P4.1 have distinct binding preferences for lipids dependent on acyl chain length and position on the glycerol backbone. Unexpectedly, the channel can also discriminate the fatty acid linkage at the sn-1 position. Of the 33 lipids interrogated using native mass spectrometry, phosphatidic acid (PA) had the lowest equilibrium dissociation constants for both isoforms of K2P4.1. Liposome potassium flux assays with K2P4.1 reconstituted in defined lipid environments show that those containing PA activate the channel in a dose-dependent fashion. Our results begin to define the molecular requirements for the specific binding of lipids to K2P4.1.
Collapse
|
30
|
Tu N, Liang D, Zhang P. Whole-exome sequencing and genome-wide evolutionary analyses identify novel candidate genes associated with infrared perception in pit vipers. Sci Rep 2020; 10:13033. [PMID: 32747674 PMCID: PMC7400743 DOI: 10.1038/s41598-020-69843-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Pit vipers possess a unique thermal sensory system consisting of facial pits that allow them to detect minute temperature fluctuations within their environments. Biologists have long attempted to elucidate the genetic basis underlying the infrared perception of pit vipers. Early studies have shown that the TRPA1 gene is the thermal sensor associated with infrared detection in pit vipers. However, whether genes other than TRPA1 are also involved in the infrared perception of pit vipers remains unknown. Here, we sequenced the whole exomes of ten snake species and performed genome-wide evolutionary analyses to search for novel candidate genes that might be involved in the infrared perception of pit vipers. We applied both branch-length-comparison and selection-pressure-alteration analyses to identify genes that specifically underwent accelerated evolution in the ancestral lineage of pit vipers. A total of 47 genes were identified. These genes were significantly enriched in the ion transmembrane transporter, stabilization of membrane potential, and temperature gating activity functional categories. The expression levels of these candidate genes in relevant nerve tissues (trigeminal ganglion, dorsal root ganglion, midbrain, and cerebrum) were also investigated in this study. We further chose one of our candidate genes, the potassium channel gene KCNK4, as an example to discuss its possible role in the infrared perception of pit vipers. Our study provides the first genome-wide survey of infrared perception-related genes in pit vipers via comparative evolutionary analyses and reveals valuable candidate genes for future functional studies.
Collapse
Affiliation(s)
- Na Tu
- State Key Laboratory of Biocontrol, College of Ecology and Evolution, School of Life Sciences, Higher Education Mega Center, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Dan Liang
- State Key Laboratory of Biocontrol, College of Ecology and Evolution, School of Life Sciences, Higher Education Mega Center, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Peng Zhang
- State Key Laboratory of Biocontrol, College of Ecology and Evolution, School of Life Sciences, Higher Education Mega Center, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
31
|
Zou X, Conrad LJ, Koschinsky K, Schlichthörl G, Preisig-Müller R, Netz E, Krüger J, Daut J, Renigunta V. The Phosphodiesterase Inhibitor IBMX Blocks the Potassium Channel THIK-1 from the Extracellular Side. Mol Pharmacol 2020; 98:143-155. [PMID: 32616523 DOI: 10.1124/molpharm.120.000011] [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: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022] Open
Abstract
The two-pore domain potassium channel (K2P-channel) THIK-1 has several predicted protein kinase A (PKA) phosphorylation sites. In trying to elucidate whether THIK-1 is regulated via PKA, we expressed THIK-1 channels in a mammalian cell line (CHO cells) and used the phosphodiesterase inhibitor 3-isobutyl-1-methyl-xanthine (IBMX) as a pharmacological tool to induce activation of PKA. Using the whole-cell patch-clamp recording, we found that THIK-1 currents were inhibited by application of IBMX with an IC50 of 120 µM. Surprisingly, intracellular application of IBMX or of the second messenger cAMP via the patch pipette had no effect on THIK-1 currents. In contrast, extracellular application of IBMX produced a rapid and reversible inhibition of THIK-1. In patch-clamp experiments with outside-out patches, THIK-1 currents were also inhibited by extracellular application of IBMX. Expression of THIK-1 channels in Xenopus oocytes was used to compare wild-type channels with mutated channels. Mutation of the putative PKA phosphorylation sites did not change the inhibitory effect of IBMX on THIK-1 currents. Mutational analysis of all residues of the (extracellular) helical cap of THIK-1 showed that mutation of the arginine residue at position 92, which is in the linker between cap helix 2 and pore helix 1, markedly reduced the inhibitory effect of IBMX. This flexible linker region, which is unique for each K2P-channel subtype, may be a possible target of channel-specific blockers. SIGNIFICANCE STATEMENT: The potassium channel THIK-1 is strongly expressed in the central nervous system. We studied the effect of 3-isobutyl-1-methyl-xanthine (IBMX) on THIK-1 currents. IBMX inhibits breakdown of cAMP and thus activates protein kinase A (PKA). Surprisingly, THIK-1 current was inhibited when IBMX was applied from the extracellular side of the membrane, but not from the intracellular side. Our results suggest that IBMX binds directly to the channel and that the inhibition of THIK-1 current was not related to activation of PKA.
Collapse
Affiliation(s)
- Xinle Zou
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Linus J Conrad
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Kristin Koschinsky
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Günter Schlichthörl
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Regina Preisig-Müller
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Eugen Netz
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Jens Krüger
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Jürgen Daut
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| | - Vijay Renigunta
- Institute of Physiology and Pathophysiology, Marburg University, Marburg, Germany (X.Z., L.J.C., K.K., G.S., R.P.-M., J.D., V.R.); Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany (E.N.); and High Performance and Cloud Computing Group, IT Center, University of Tübingen, Tübingen, Germany (J.K.)
| |
Collapse
|
32
|
Lee AG. Interfacial Binding Sites for Cholesterol on Kir, Kv, K 2P, and Related Potassium Channels. Biophys J 2020; 119:35-47. [PMID: 32553129 PMCID: PMC7335934 DOI: 10.1016/j.bpj.2020.05.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 12/30/2022] Open
Abstract
Inwardly rectifying, voltage-gated, two-pore domain, and related K+ channels are located in eukaryotic membranes rich in cholesterol. Here, molecular docking is used to detect specific binding sites ("hot spots") for cholesterol on K+ channels with characteristics that match those of known cholesterol binding sites. The transmembrane surfaces of all available high-resolution structures for K+ channels were swept for potential binding sites. Cholesterol poses were found to be located largely in hollows between protein ridges. A comparison between cholesterol poses and resolved phospholipids suggests that not all cholesterol molecules binding to the transmembrane surface of a K+ channel will result in displacement of a phospholipid molecule from the surface. Competition between cholesterol binding and binding of anionic phospholipids essential for activity could explain some of the effects of cholesterol on channel function.
Collapse
Affiliation(s)
- Anthony G Lee
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom.
| |
Collapse
|
33
|
Wiedmann F, Rinné S, Donner B, Decher N, Katus HA, Schmidt C. Mechanosensitive TREK-1 two-pore-domain potassium (K 2P) channels in the cardiovascular system. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:126-135. [PMID: 32553901 DOI: 10.1016/j.pbiomolbio.2020.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/01/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
TWIK-related K+ channel (TREK-1) two-pore-domain potassium (K2P) channels mediate background potassium currents and regulate cellular excitability in many different types of cells. Their functional activity is controlled by a broad variety of different physiological stimuli, such as temperature, extracellular or intracellular pH, lipids and mechanical stress. By linking cellular excitability to mechanical stress, TREK-1 currents might be important to mediate parts of the mechanoelectrical feedback described in the heart. Furthermore, TREK-1 currents might contribute to the dysregulation of excitability in the heart in pathophysiological situations, such as those caused by abnormal stretch or ischaemia-associated cell swelling, thereby contributing to arrhythmogenesis. In this review, we focus on the functional role of TREK-1 in the heart and its putative contribution to cardiac mechanoelectrical coupling. Its cardiac expression among different species is discussed, alongside with functional evidence for TREK-1 currents in cardiomyocytes. In addition, evidence for the involvement of TREK-1 currents in different cardiac arrhythmias, such as atrial fibrillation or ventricular tachycardia, is summarized. Furthermore, the role of TREK-1 and its interaction partners in the regulation of the cardiac heart rate is reviewed. Finally, we focus on the significance of TREK-1 in the development of cardiac hypertrophy, cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Felix Wiedmann
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany; HCR, Heidelberg Center for Heart Rhythm Disorders, University Hospital Heidelberg, Heidelberg, Germany
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - Philipps-University Marburg, Marburg, Germany
| | - Birgit Donner
- Pediatric Cardiology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - Philipps-University Marburg, Marburg, Germany
| | - Hugo A Katus
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany; HCR, Heidelberg Center for Heart Rhythm Disorders, University Hospital Heidelberg, Heidelberg, Germany
| | - Constanze Schmidt
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany; HCR, Heidelberg Center for Heart Rhythm Disorders, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
34
|
Lewis A, McCrossan ZA, Manville RW, Popa MO, Cuello LG, Goldstein SAN. TOK channels use the two gates in classical K + channels to achieve outward rectification. FASEB J 2020; 34:8902-8919. [PMID: 32519783 DOI: 10.1096/fj.202000545r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 11/11/2022]
Abstract
TOKs are outwardly rectifying K+ channels in fungi with two pore-loops and eight transmembrane spans. Here, we describe the TOKs from four pathogens that cause the majority of life-threatening fungal infections in humans. These TOKs pass large currents only in the outward direction like the canonical isolate from Saccharomyces cerevisiae (ScTOK), and distinct from other K+ channels. ScTOK, AfTOK1 (Aspergillus fumigatus), and H99TOK (Cryptococcus neoformans grubii) are K+ -selective and pass current above the K+ reversal potential. CaTOK (Candida albicans) and CnTOK (Cryptococcus neoformans neoformans) pass both K+ and Na+ and conduct above a reversal potential reflecting the mixed permeability of their selectivity filter. Mutations in CaTOK and ScTOK at sites homologous to those that open the internal gates in classical K+ channels are shown to produce inward TOK currents. A favored model for outward rectification is proposed whereby the reversal potential determines ion occupancy, and thus, conductivity, of the selectivity filter gate that is coupled to an imperfectly restrictive internal gate, permitting the filter to sample ion concentrations on both sides of the membrane.
Collapse
Affiliation(s)
- Anthony Lewis
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Zoe A McCrossan
- NIHR Evaluation, Trials and Studies Coordinating Centre (NETSCC), University of Southampton, Southampton, UK
| | - Rían W Manville
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - M Oana Popa
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Luis G Cuello
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Steve A N Goldstein
- Departments of Physiology & Biophysics and Pediatrics, School of Medicine, Samueli College of Health Sciences, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
35
|
A lower X-gate in TASK channels traps inhibitors within the vestibule. Nature 2020; 582:443-447. [PMID: 32499642 DOI: 10.1038/s41586-020-2250-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/12/2020] [Indexed: 12/23/2022]
Abstract
TWIK-related acid-sensitive potassium (TASK) channels-members of the two pore domain potassium (K2P) channel family-are found in neurons1, cardiomyocytes2-4 and vascular smooth muscle cells5, where they are involved in the regulation of heart rate6, pulmonary artery tone5,7, sleep/wake cycles8 and responses to volatile anaesthetics8-11. K2P channels regulate the resting membrane potential, providing background K+ currents controlled by numerous physiological stimuli12-15. Unlike other K2P channels, TASK channels are able to bind inhibitors with high affinity, exceptional selectivity and very slow compound washout rates. As such, these channels are attractive drug targets, and TASK-1 inhibitors are currently in clinical trials for obstructive sleep apnoea and atrial fibrillation16. In general, potassium channels have an intramembrane vestibule with a selectivity filter situated above and a gate with four parallel helices located below; however, the K2P channels studied so far all lack a lower gate. Here we present the X-ray crystal structure of TASK-1, and show that it contains a lower gate-which we designate as an 'X-gate'-created by interaction of the two crossed C-terminal M4 transmembrane helices at the vestibule entrance. This structure is formed by six residues (243VLRFMT248) that are essential for responses to volatile anaesthetics10, neurotransmitters13 and G-protein-coupled receptors13. Mutations within the X-gate and the surrounding regions markedly affect both the channel-open probability and the activation of the channel by anaesthetics. Structures of TASK-1 bound to two high-affinity inhibitors show that both compounds bind below the selectivity filter and are trapped in the vestibule by the X-gate, which explains their exceptionally low washout rates. The presence of the X-gate in TASK channels explains many aspects of their physiological and pharmacological behaviour, which will be beneficial for the future development and optimization of TASK modulators for the treatment of heart, lung and sleep disorders.
Collapse
|
36
|
Qiu Y, Huang L, Fu J, Han C, Fang J, Liao P, Chen Z, Mo Y, Sun P, Liao D, Yang L, Wang J, Zhang Q, Liu J, Liu F, Liu T, Huang W, Yang H, Jiang R. TREK Channel Family Activator with a Well-Defined Structure–Activation Relationship for Pain and Neurogenic Inflammation. J Med Chem 2020; 63:3665-3677. [PMID: 32162512 DOI: 10.1021/acs.jmedchem.9b02163] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yunguang Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Jie Fu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Chenxia Han
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Jing Fang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Zhuo Chen
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Yiqing Mo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Peihua Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| | - Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tingting Liu
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, Chengdu 610000, China
| |
Collapse
|
37
|
Bustos D, Bedoya M, Ramírez D, Concha G, Zúñiga L, Decher N, Hernández-Rodríguez EW, Sepúlveda FV, Martínez L, González W. Elucidating the Structural Basis of the Intracellular pH Sensing Mechanism of TASK-2 K 2P Channels. Int J Mol Sci 2020; 21:ijms21020532. [PMID: 31947679 PMCID: PMC7013731 DOI: 10.3390/ijms21020532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/25/2019] [Accepted: 01/08/2020] [Indexed: 11/23/2022] Open
Abstract
Two-pore domain potassium (K2P) channels maintain the cell’s background conductance by stabilizing the resting membrane potential. They assemble as dimers possessing four transmembrane helices in each subunit. K2P channels were crystallized in “up” and “down” states. The movements of the pore-lining transmembrane TM4 helix produce the aperture or closure of side fenestrations that connect the lipid membrane with the central cavity. When the TM4 helix is in the up-state, the fenestrations are closed, while they are open in the down-state. It is thought that the fenestration states are related to the activity of K2P channels and the opening of the channels preferentially occurs from the up-state. TASK-2, a member of the TALK subfamily of K2P channels, is opened by intracellular alkalization leading the deprotonation of the K245 residue at the end of the TM4 helix. This charge neutralization of K245 could be sensitive or coupled to the fenestration state. Here, we describe the relationship between the states of the intramembrane fenestrations and K245 residue in TASK-2 channel. By using molecular modeling and simulations, we show that the protonated state of K245 (K245+) favors the open fenestration state and, symmetrically, that the open fenestration state favors the protonated state of the lysine residue. We show that the channel can be completely blocked by Prozac, which is known to induce fenestration opening in TREK-2. K245 protonation and fenestration aperture have an additive effect on the conductance of the channel. The opening of the fenestrations with K245+ increases the entrance of lipids into the selectivity filter, blocking the channel. At the same time, the protonation of K245 introduces electrostatic potential energy barriers to ion entrance. We computed the free energy profiles of ion penetration into the channel in different fenestration and K245 protonation states, to show that the effects of the two transformations are summed up, leading to maximum channel blocking. Estimated rates of ion transport are in qualitative agreement with experimental results and support the hypothesis that the most important barrier for ion transport under K245+ and open fenestration conditions is the entrance of the ions into the channel.
Collapse
Affiliation(s)
- Daniel Bustos
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca 3460000, Chile; (D.B.); (M.B.)
- Departamento de Computación e Industrias, Facultad de Ciencias de la Ingeniería, Universidad Católica del Maule, Talca 3460000, Chile
| | - Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca 3460000, Chile; (D.B.); (M.B.)
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago 8380453, Chile;
| | - Guierdy Concha
- Centro de Investigaciones Médicas, Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile; (G.C.); (L.Z.)
- Magíster en Gestión de Operaciones, Facultad de Ingeniería (Campus Los Niches), Universidad de Talca, Talca 3460000, Chile
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas, Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile; (G.C.); (L.Z.)
- Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca 3460000, Chile
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, University of Marburg, D-35037 Marburg, Germany;
| | | | - Francisco V. Sepúlveda
- Centro de Estudios Científicos (CECs), Avenida Arturo Prat 514, Valdivia 5110466, Chile
- Correspondence: (F.V.S.); (L.M.); (W.G.)
| | - Leandro Martínez
- Institute of Chemistry and Center for Computing in Engineering & Science, University of Campinas, Campinas 13083-861 SP, Brazil
- Correspondence: (F.V.S.); (L.M.); (W.G.)
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca 3460000, Chile; (D.B.); (M.B.)
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca 3460000, Chile
- Correspondence: (F.V.S.); (L.M.); (W.G.)
| |
Collapse
|
38
|
Kopec W, Rothberg BS, de Groot BL. Molecular mechanism of a potassium channel gating through activation gate-selectivity filter coupling. Nat Commun 2019; 10:5366. [PMID: 31772184 PMCID: PMC6879586 DOI: 10.1038/s41467-019-13227-w] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023] Open
Abstract
Potassium channels are presumed to have two allosterically coupled gates, the activation gate and the selectivity filter gate, that control channel opening, closing, and inactivation. However, the molecular mechanism of how these gates regulate K+ ion flow through the channel remains poorly understood. An activation process, occurring at the selectivity filter, has been recently proposed for several potassium channels. Here, we use X-ray crystallography and extensive molecular dynamics simulations, to study ion permeation through a potassium channel MthK, for various opening levels of both gates. We find that the channel conductance is controlled at the selectivity filter, whose conformation depends on the activation gate. The crosstalk between the gates is mediated through a collective motion of channel helices, involving hydrophobic contacts between an isoleucine and a conserved threonine in the selectivity filter. We propose a gating model of selectivity filter-activated potassium channels, including pharmacologically relevant two-pore domain (K2P) and big potassium (BK) channels.
Collapse
Affiliation(s)
- Wojciech Kopec
- Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.
| | - Brad S Rothberg
- Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA, 19140, USA
| | - Bert L de Groot
- Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.
| |
Collapse
|
39
|
Brohawn SG, Wang W, Handler A, Campbell EB, Schwarz JR, MacKinnon R. The mechanosensitive ion channel TRAAK is localized to the mammalian node of Ranvier. eLife 2019; 8:50403. [PMID: 31674909 PMCID: PMC6824864 DOI: 10.7554/elife.50403] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
TRAAK is a membrane tension-activated K+ channel that has been associated through behavioral studies to mechanical nociception. We used specific monoclonal antibodies in mice to show that TRAAK is localized exclusively to nodes of Ranvier, the action potential propagating elements of myelinated nerve fibers. Approximately 80 percent of myelinated nerve fibers throughout the central and peripheral nervous system contain TRAAK in what is likely an all-nodes or no-nodes per axon fashion. TRAAK is not observed at the axon initial segment where action potentials are first generated. We used polyclonal antibodies, the TRAAK inhibitor RU2 and node clamp amplifiers to demonstrate the presence and functional properties of TRAAK in rat nerve fibers. TRAAK contributes to the ‘leak’ K+ current in mammalian nerve fiber conduction by hyperpolarizing the resting membrane potential, thereby increasing Na+ channel availability for action potential propagation. We speculate on why nodes of Ranvier contain a mechanosensitive K+ channel.
Collapse
Affiliation(s)
- Stephen G Brohawn
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Weiwei Wang
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Annie Handler
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Ernest B Campbell
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Jürgen R Schwarz
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
40
|
Discovery of Novel TASK-3 Channel Blockers Using a Pharmacophore-Based Virtual Screening. Int J Mol Sci 2019; 20:ijms20164014. [PMID: 31426491 PMCID: PMC6720600 DOI: 10.3390/ijms20164014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
TASK-3 is a two-pore domain potassium (K2P) channel highly expressed in the hippocampus, cerebellum, and cortex. TASK-3 has been identified as an oncogenic potassium channel and it is overexpressed in different cancer types. For this reason, the development of new TASK-3 blockers could influence the pharmacological treatment of cancer and several neurological conditions. In the present work, we searched for novel TASK-3 blockers by using a virtual screening protocol that includes pharmacophore modeling, molecular docking, and free energy calculations. With this protocol, 19 potential TASK-3 blockers were identified. These molecules were tested in TASK-3 using patch clamp, and one blocker (DR16) was identified with an IC50 = 56.8 ± 3.9 μM. Using DR16 as a scaffold, we designed DR16.1, a novel TASK-3 inhibitor, with an IC50 = 14.2 ± 3.4 μM. Our finding takes on greater relevance considering that not many inhibitory TASK-3 modulators have been reported in the scientific literature until today. These two novel TASK-3 channel inhibitors (DR16 and DR16.1) are the first compounds found using a pharmacophore-based virtual screening and rational drug design protocol.
Collapse
|
41
|
Bedoya M, Rinné S, Kiper AK, Decher N, González W, Ramírez D. TASK Channels Pharmacology: New Challenges in Drug Design. J Med Chem 2019; 62:10044-10058. [PMID: 31260312 DOI: 10.1021/acs.jmedchem.9b00248] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Rational drug design targeting ion channels is an exciting and always evolving research field. New medicinal chemistry strategies are being implemented to explore the wild chemical space and unravel the molecular basis of the ion channels modulators binding mechanisms. TASK channels belong to the two-pore domain potassium channel family and are modulated by extracellular acidosis. They are extensively distributed along the cardiovascular and central nervous systems, and their expression is up- and downregulated in different cancer types, which makes them an attractive therapeutic target. However, TASK channels remain unexplored, and drugs designed to target these channels are poorly selective. Here, we review TASK channels properties and their known blockers and activators, considering the new challenges in ion channels drug design and focusing on the implementation of computational methodologies in the drug discovery process.
Collapse
Affiliation(s)
- Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior, MCMBB , Philipps-University of Marburg , Deutschhausstraße 2 , Marburg 35037 , Germany
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Talca , 1 Poniente No. 1141 , 3460000 Talca , Chile
| | - David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , El Llano Subercaseaux 2801, Piso 6 , 8900000 Santiago , Chile
| |
Collapse
|
42
|
Şterbuleac D. Molecular determinants of chemical modulation of two-pore domain potassium channels. Chem Biol Drug Des 2019; 94:1596-1614. [PMID: 31124599 DOI: 10.1111/cbdd.13571] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 05/05/2019] [Indexed: 12/16/2022]
Abstract
The K+ ion channels comprising the two-pore domain (K2P) family have specific biophysical roles in generating the critical regulatory K+ current. Ion flow through K2P channels and, implicitly, channel regulation is mediated by diverse metabolic and physical inputs such as mechanical stimulation, interaction with lipids or endogenous regulators, intra- or extracellular pH, and phosphorylation, while their function can be finely tuned by chemical compounds. In the latter category, some drug-channel interactions can lead to side effects or have clinical action, while identifying novel chemical modulators of K2Ps is an area of intense research. Due to their cellular and therapeutic importance, much attention was turned to these channels in recent years and several experimental approaches have pinpointed the molecular determinants of K2P chemical modulation. Given their unique structural features and properties, chemical modulators act on K2P channels in multiple and diverse ways. In this review, the particularities of K2P modulation by chemical compounds, such as binding modality, affinity, or position, are identified, synthesized, and linked to structural and functional properties in order to refer to how activators and blockers modify channel function and vice versa, focusing on specificity related to protein structure (and its modification) and cross-linking information among different subfamilies.
Collapse
Affiliation(s)
- Daniel Şterbuleac
- Doctoral School of Biology, Faculty of Biology, "Alexandru Ioan Cuza" University of Iasi, Iasi, Romania
| |
Collapse
|
43
|
Ramírez D, Bedoya M, Kiper AK, Rinné S, Morales-Navarro S, Hernández-Rodríguez EW, Sepúlveda FV, Decher N, González W. Structure/Activity Analysis of TASK-3 Channel Antagonists Based on a 5,6,7,8 tetrahydropyrido[4,3-d]pyrimidine. Int J Mol Sci 2019; 20:ijms20092252. [PMID: 31067753 PMCID: PMC6539479 DOI: 10.3390/ijms20092252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/18/2019] [Accepted: 04/20/2019] [Indexed: 11/16/2022] Open
Abstract
TASK-3 potassium (K+) channels are highly expressed in the central nervous system, regulating the membrane potential of excitable cells. TASK-3 is involved in neurotransmitter action and has been identified as an oncogenic K+ channel. For this reason, the understanding of the action mechanism of pharmacological modulators of these channels is essential to obtain new therapeutic strategies. In this study we describe the binding mode of the potent antagonist PK-THPP into the TASK-3 channel. PK-THPP blocks TASK-1, the closest relative channel of TASK-3, with almost nine-times less potency. Our results confirm that the binding is influenced by the fenestrations state of TASK-3 channels and occurs when they are open. The binding is mainly governed by hydrophobic contacts between the blocker and the residues of the binding site. These interactions occur not only for PK-THPP, but also for the antagonist series based on 5,6,7,8 tetrahydropyrido[4,3-d]pyrimidine scaffold (THPP series). However, the marked difference in the potency of THPP series compounds such as 20b, 21, 22 and 23 (PK-THPP) respect to compounds such as 17b, inhibiting TASK-3 channels in the micromolar range is due to the presence of a hydrogen bond acceptor group that can establish interactions with the threonines of the selectivity filter.
Collapse
Affiliation(s)
- David Ramírez
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile. El Llano Subercaseaux 2801-Piso 6, 7500912 Santiago, Chile.
| | - Mauricio Bedoya
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca. 1 Poniente No. 1141, 3460000 Talca, Chile.
| | - Aytug K Kiper
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 2, 35037 Marburg, Germany.
| | - Susanne Rinné
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 2, 35037 Marburg, Germany.
| | - Samuel Morales-Navarro
- Bachillerato en Ciencias, Facultad de Ciencias, Universidad Santo Tomás, Av. Circunvalación Poniente #1855, 3460000 Talca, Chile.
| | - Erix W Hernández-Rodríguez
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca. 1 Poniente No. 1141, 3460000 Talca, Chile.
- Escuela de Química y Farmacia. Facultad de Medicina. Universidad Católica del Maule, 3460000 Talca, Chile.
| | | | - Niels Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, Deutschhausstraße 2, 35037 Marburg, Germany.
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca. 1 Poniente No. 1141, 3460000 Talca, Chile.
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, 3460000 Talca, Chile.
| |
Collapse
|
44
|
Schewe M, Sun H, Mert Ü, Mackenzie A, Pike ACW, Schulz F, Constantin C, Vowinkel KS, Conrad LJ, Kiper AK, Gonzalez W, Musinszki M, Tegtmeier M, Pryde DC, Belabed H, Nazare M, de Groot BL, Decher N, Fakler B, Carpenter EP, Tucker SJ, Baukrowitz T. A pharmacological master key mechanism that unlocks the selectivity filter gate in K + channels. Science 2019; 363:875-880. [PMID: 30792303 PMCID: PMC6982535 DOI: 10.1126/science.aav0569] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Potassium (K+) channels have been evolutionarily tuned for activation by diverse biological stimuli, and pharmacological activation is thought to target these specific gating mechanisms. Here we report a class of negatively charged activators (NCAs) that bypass the specific mechanisms but act as master keys to open K+ channels gated at their selectivity filter (SF), including many two-pore domain K+ (K2P) channels, voltage-gated hERG (human ether-à-go-go-related gene) channels and calcium (Ca2+)-activated big-conductance potassium (BK)-type channels. Functional analysis, x-ray crystallography, and molecular dynamics simulations revealed that the NCAs bind to similar sites below the SF, increase pore and SF K+ occupancy, and open the filter gate. These results uncover an unrecognized polypharmacology among K+ channel activators and highlight a filter gating machinery that is conserved across different families of K+ channels with implications for rational drug design.
Collapse
Affiliation(s)
- Marcus Schewe
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany.
| | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department of Structural Biology, 13125 Berlin, Germany
| | - Ümit Mert
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - Alexandra Mackenzie
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PN, UK
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, UK
| | - Ashley C W Pike
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
| | - Friederike Schulz
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - Cristina Constantin
- Institute of Physiology II, Albert-Ludwigs University of Freiburg, 79104 Freiburg, Germany
- Centers for Biological Signaling Studies CIBSS and BIOSS, 79104 Freiburg, Germany
| | - Kirsty S Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 35037 Marburg, Germany
| | - Linus J Conrad
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PN, UK
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, UK
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 35037 Marburg, Germany
| | - Wendy Gonzalez
- Centro de Bioinformatica y Simulacion Molecular, Universidad de Talca, 3465548 Talca, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, 3465548 Talca, Chile
| | - Marianne Musinszki
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - Marie Tegtmeier
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - David C Pryde
- Pfizer Worldwide Medicinal Chemistry, Neuroscience and Pain Research Unit, Portway Building, Granta Park, Great Abington, Cambridgeshire CB21 6GS, UK
| | - Hassane Belabed
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department of Medicinal Chemistry, 13125 Berlin, Germany
| | - Marc Nazare
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Department of Medicinal Chemistry, 13125 Berlin, Germany
| | - Bert L de Groot
- Computational Biomolecular Dynamics Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps-University of Marburg, 35037 Marburg, Germany
| | - Bernd Fakler
- Institute of Physiology II, Albert-Ludwigs University of Freiburg, 79104 Freiburg, Germany
- Centers for Biological Signaling Studies CIBSS and BIOSS, 79104 Freiburg, Germany
| | - Elisabeth P Carpenter
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, UK
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PN, UK
| | - Stephen J Tucker
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PN, UK
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, UK
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts University of Kiel, 24118 Kiel, Germany.
| |
Collapse
|
45
|
Pope L, Arrigoni C, Lou H, Bryant C, Gallardo-Godoy A, Renslo AR, Minor DL. Protein and Chemical Determinants of BL-1249 Action and Selectivity for K 2P Channels. ACS Chem Neurosci 2018; 9:3153-3165. [PMID: 30089357 PMCID: PMC6302903 DOI: 10.1021/acschemneuro.8b00337] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
K2P potassium channels generate leak currents that stabilize the resting membrane potential of excitable cells. Various K2P channels are implicated in pain, ischemia, depression, migraine, and anesthetic responses, making this family an attractive target for small molecule modulator development efforts. BL-1249, a compound from the fenamate class of nonsteroidal anti-inflammatory drugs is known to activate K2P2.1(TREK-1), the founding member of the thermo- and mechanosensitive TREK subfamily; however, its mechanism of action and effects on other K2P channels are not well-defined. Here, we demonstrate that BL-1249 extracellular application activates all TREK subfamily members but has no effect on other K2P subfamilies. Patch clamp experiments demonstrate that, similar to the diverse range of other chemical and physical TREK subfamily gating cues, BL-1249 stimulates the selectivity filter "C-type" gate that controls K2P function. BL-1249 displays selectivity among the TREK subfamily, activating K2P2.1(TREK-1) and K2P10.1(TREK-2) ∼10-fold more potently than K2P4.1(TRAAK). Investigation of mutants and K2P2.1(TREK-1)/K2P4.1(TRAAK) chimeras highlight the key roles of the C-terminal tail in BL-1249 action and identify the M2/M3 transmembrane helix interface as a key site of BL-1249 selectivity. Synthesis and characterization of a set of BL-1249 analogs demonstrates that both the tetrazole and opposing tetralin moieties are critical for function, whereas the conformational mobility between the two ring systems impacts selectivity. Together, our findings underscore the landscape of modes by which small molecules can affect K2P channels and provide crucial information for the development of better and more selective K2P modulators of the TREK subfamily.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel L. Minor
- Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 United States
| |
Collapse
|
46
|
Mutations in KCNK4 that Affect Gating Cause a Recognizable Neurodevelopmental Syndrome. Am J Hum Genet 2018; 103:621-630. [PMID: 30290154 DOI: 10.1016/j.ajhg.2018.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/31/2018] [Indexed: 12/15/2022] Open
Abstract
Aberrant activation or inhibition of potassium (K+) currents across the plasma membrane of cells has been causally linked to altered neurotransmission, cardiac arrhythmias, endocrine dysfunction, and (more rarely) perturbed developmental processes. The K+ channel subfamily K member 4 (KCNK4), also known as TRAAK (TWIK-related arachidonic acid-stimulated K+ channel), belongs to the mechano-gated ion channels of the TRAAK/TREK subfamily of two-pore-domain (K2P) K+ channels. While K2P channels are well known to contribute to the resting membrane potential and cellular excitability, their involvement in pathophysiological processes remains largely uncharacterized. We report that de novo missense mutations in KCNK4 cause a recognizable syndrome with a distinctive facial gestalt, for which we propose the acronym FHEIG (facial dysmorphism, hypertrichosis, epilepsy, intellectual disability/developmental delay, and gingival overgrowth). Patch-clamp analyses documented a significant gain of function of the identified KCNK4 channel mutants basally and impaired sensitivity to mechanical stimulation and arachidonic acid. Co-expression experiments indicated a dominant behavior of the disease-causing mutations. Molecular dynamics simulations consistently indicated that mutations favor sealing of the lateral intramembrane fenestration that has been proposed to negatively control K+ flow by allowing lipid access to the central cavity of the channel. Overall, our findings illustrate the pleiotropic effect of dysregulated KCNK4 function and provide support to the hypothesis of a gating mechanism based on the lateral fenestrations of K2P channels.
Collapse
|
47
|
Concha G, Bustos D, Zúñiga R, Catalán MA, Zúñiga L. The Insensitivity of TASK-3 K₂P Channels to External Tetraethylammonium (TEA) Partially Depends on the Cap Structure. Int J Mol Sci 2018; 19:ijms19082437. [PMID: 30126179 PMCID: PMC6121469 DOI: 10.3390/ijms19082437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/12/2023] Open
Abstract
Two-pore domain K⁺ channels (K₂P) display a characteristic extracellular cap structure formed by two M1-P1 linkers, the functional role of which is poorly understood. It has been proposed that the presence of the cap explains the insensitivity of K₂P channels to several K⁺ channel blockers including tetraethylammonium (TEA). We have explored this hypothesis using mutagenesis and functional analysis, followed by molecular simulations. Our results show that the deletion of the cap structure of TASK-3 (TWIK-related acid-sensitive K⁺ channel) generates a TEA-sensitive channel with an IC50 of 11.8 ± 0.4 mM. The enhanced sensitivity to TEA displayed by the cap-less channel is also explained by the presence of an extra tyrosine residue at position 99. These results were corroborated by molecular simulation analysis, which shows an increased stability in the binding of TEA to the cap-less channel when a ring of four tyrosine is present at the external entrance of the permeation pathway. Consistently, Y99A or Y205A single-residue mutants generated in a cap-less channel backbone resulted in TASK-3 channels with low affinity to external TEA.
Collapse
Affiliation(s)
- Guierdy Concha
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Daniel Bustos
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Rafael Zúñiga
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| | - Marcelo A Catalán
- Laboratorio de Fisiología Epitelial, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1130000, Chile.
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas (CIM), Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Escuela de Medicina, Universidad de Talca, Talca 3460000, Chile.
| |
Collapse
|
48
|
Falzone ME, Malvezzi M, Lee BC, Accardi A. Known structures and unknown mechanisms of TMEM16 scramblases and channels. J Gen Physiol 2018; 150:933-947. [PMID: 29915161 PMCID: PMC6028493 DOI: 10.1085/jgp.201711957] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Falzone et al. interpret the mechanisms underlying the activity of TMEM16 family members from recent structural and functional work. The TMEM16 family of membrane proteins is composed of both Ca2+-gated Cl− channels and Ca2+-dependent phospholipid scramblases. The functional diversity of TMEM16s underlies their involvement in numerous signal transduction pathways that connect changes in cytosolic Ca2+ levels to cellular signaling networks. Indeed, defects in the function of several TMEM16s cause a variety of genetic disorders, highlighting their fundamental pathophysiological importance. Here, we review how our mechanistic understanding of TMEM16 function has been shaped by recent functional and structural work. Remarkably, the recent determination of near-atomic-resolution structures of TMEM16 proteins of both functional persuasions has revealed how relatively minimal rearrangements in the substrate translocation pathway are sufficient to precipitate the dramatic functional differences that characterize the family. These structures, when interpreted in the light of extensive functional analysis, point to an unusual mechanism for Ca2+-dependent activation of TMEM16 proteins in which substrate permeation is regulated by a combination of conformational rearrangements and electrostatics. These breakthroughs pave the way to elucidate the mechanistic bases of ion and lipid transport by the TMEM16 proteins and unravel the molecular links between these transport activities and their function in human pathophysiology.
Collapse
Affiliation(s)
- Maria E Falzone
- Department of Biochemistry, Weill Cornell Medical School, New York, NY
| | - Mattia Malvezzi
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Byoung-Cheol Lee
- Department of Anesthesiology, Weill Cornell Medical School, New York, NY
| | - Alessio Accardi
- Department of Biochemistry, Weill Cornell Medical School, New York, NY .,Department of Anesthesiology, Weill Cornell Medical School, New York, NY.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medical School, New York, NY
| |
Collapse
|
49
|
Navarro-Retamal C, Caballero J. Energetic differences between non-domain-swapped and domain-swapped chain connectivities in the K2P potassium channel TRAAK. RSC Adv 2018; 8:26610-26618. [PMID: 35541058 PMCID: PMC9083029 DOI: 10.1039/c8ra04159h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/19/2018] [Indexed: 01/15/2023] Open
Abstract
Two-pore domain (K2P) channels are twofold symmetric K+ channels which control cell excitability by enabling the leak of potassium ions from cells in response to physicochemical stimuli. Crystallization of K2P channels revealed the presence of several structural features, which include an external cap. In the available crystallographic structures, the cap is present as non-domain-swapped (NDS) and domain-swapped (DS) chain conformations, where DS chain conformation exchanges two opposing outer helices 180° around the channel. In this work, energy differences between the residues located at the highest point of the cap in NDS and DS conformations were evaluated for TRAAK, a K2P channel that was crystallized in both conformations. Results indicated a preference for DS conformation, but this result is not extensible to TASK K2P channels. In the available crystallographic structures of K2P channels, the cap is present as non-domain-swapped (NDS) and domain-swapped (DS) chain conformations.![]()
Collapse
Affiliation(s)
- Carlos Navarro-Retamal
- Centro de Bioinformática y Simulación Molecular
- Facultad de Ingeniería
- Universidad de Talca
- Talca
- Chile
| | - Julio Caballero
- Centro de Bioinformática y Simulación Molecular
- Facultad de Ingeniería
- Universidad de Talca
- Talca
- Chile
| |
Collapse
|
50
|
McClenaghan C, Schewe M, Aryal P, Carpenter EP, Baukrowitz T, Tucker SJ. Polymodal activation of the TREK-2 K2P channel produces structurally distinct open states. J Gen Physiol 2017; 147:497-505. [PMID: 27241700 PMCID: PMC4886281 DOI: 10.1085/jgp.201611601] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/09/2016] [Indexed: 01/08/2023] Open
Abstract
TREK channels, which are gated open by a wide range of stimuli, exist in at least two conformations known as “up” and “down.” McClenaghan et al. show that the channel can be open in both of these conformations and that gating is primarily achieved by the channel’s selectivity filter. The TREK subfamily of two-pore domain (K2P) K+ channels exhibit polymodal gating by a wide range of physical and chemical stimuli. Crystal structures now exist for these channels in two main states referred to as the “up” and “down” conformations. However, recent studies have resulted in contradictory and mutually exclusive conclusions about the functional (i.e., conductive) status of these two conformations. To address this problem, we have used the state-dependent TREK-2 inhibitor norfluoxetine that can only bind to the down state, thereby allowing us to distinguish between these two conformations when activated by different stimuli. Our results reconcile these previously contradictory gating models by demonstrating that activation by pressure, temperature, voltage, and pH produce more than one structurally distinct open state and reveal that channel activation does not simply involve switching between the up and down conformations. These results also highlight the diversity of structural mechanisms that K2P channels use to integrate polymodal gating signals.
Collapse
Affiliation(s)
- Conor McClenaghan
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, England, UK OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PU, England, UK
| | - Marcus Schewe
- Department of Physiology, University of Kiel, 24118 Kiel, Germany
| | - Prafulla Aryal
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, England, UK OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PU, England, UK
| | - Elisabeth P Carpenter
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PU, England, UK Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, England, UK
| | | | - Stephen J Tucker
- Clarendon Laboratory, Department of Physics, University of Oxford, Oxford OX1 3PU, England, UK OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford OX1 3PU, England, UK
| |
Collapse
|