1
|
Yashiro K, Iwaki Y, Urata H, Kokubo M, Mori T, Sekioka Y, Isami K, Kato J, Wieting J, McGowan KM, Bridges TM, Boutaud O, Engers DW, Denton JS, Kurata H, Lindsley CW. Discovery of ONO-2920632 (VU6011887): A Highly Selective and CNS Penetrant TREK-2 (TWIK-Related K+ Channel 2) Preferring Activator In Vivo Tool Compound. ACS Chem Neurosci 2025; 16:960-967. [PMID: 39981749 PMCID: PMC11887051 DOI: 10.1021/acschemneuro.5c00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Herein we describe our initial work on the K2P family of potassium ion channels with the chemical optimization and characterization of a novel series of TWIK-Related K+ Channel (TREK)-1/2 dual activators and TREK-2 preferring activators derived from a high-throughput screening hit. The exercise provided TREK activators with good CNS penetration and others with low CNS exposure to enable exploration of both central and peripheral TREK activation. From this, ONO-2920632 (VU6011887 = 19b) emerged as a reasonably potent (human Tl+; TREK-1 EC50 = 2.8 μM (95% Emax), TREK-2 EC50 = 0.30 μM (184% Emax)), first-generation CNS penetrant (rat Kp = 0.37) in vivo tool compound with selectivity versus the other K2P channels (>91-fold selective vs TASK1, TASK2, TASK3, TRAAK, TWIK2, and 31-fold selective vs TRESK) and no significant activity in a large ancillary pharmacology panel. ONO-2920632 (VU6011887) displayed robust, dose dependent efficacy when dosed orally in a mouse pain model (acetic acid writhing assay), where it was equipotent at 3 mg/kg to the assay standard indomethacin at 10 mg/kg. The therapeutic potential of TREK channel activation has long been hampered by a lack of selective, small molecule tools, and this work provides a variety of in vivo tool compounds for the community.
Collapse
Affiliation(s)
- Kentaro Yashiro
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Yuzo Iwaki
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Hirohito Urata
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Masaya Kokubo
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Takahiro Mori
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Yoko Sekioka
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Koichi Isami
- Research
Center of Neurology, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Junya Kato
- Pharmacokinetic
Research, Ono Pharmaceutical Co., Ltd, 3-1-1 Sakurai, Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Joshua Wieting
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Kevin M. McGowan
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Olivier Boutaud
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Darren W. Engers
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jerod S. Denton
- Department
of Anesthesiology, Vanderbilt University
Medical Center, Nashville, Tennessee 37232, United States
| | - Haruto Kurata
- Drug
Discovery Chemistry, Ono Pharmaceutical
Co., Ltd, 3-1-1 Sakurai,
Shimamoto, Mishima, Osaka 618-8585, Japan
| | - Craig W. Lindsley
- Warren
Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Chemistry, Vanderbilt University, Nashville Tennessee 37232, United States
- Department
of Biochemistry, Vanderbilt University, Nashville Tennessee 37232, United States
| |
Collapse
|
2
|
Manville RW, Yoshimura RF, Yeromin AV, Hogenkamp D, van der Horst J, Zavala A, Chinedu S, Arena G, Lasky E, Fisher M, Tracy CR, Othy S, Jepps TA, Cahalan MD, Abbott GW. Polymodal K + channel modulation contributes to dual analgesic and anti-inflammatory actions of traditional botanical medicines. Commun Biol 2024; 7:1059. [PMID: 39198706 PMCID: PMC11358443 DOI: 10.1038/s42003-024-06752-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: 06/06/2023] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
Pain and inflammation contribute immeasurably to reduced quality of life, yet modern analgesic and anti-inflammatory therapeutics can cause dependence and side effects. Here, we screened 1444 plant extracts, prepared primarily from native species in California and the United States Virgin Islands, against two voltage-gated K+ channels - T-cell expressed Kv1.3 and nociceptive-neuron expressed Kv7.2/7.3. A subset of extracts both inhibits Kv1.3 and activates Kv7.2/7.3 at hyperpolarized potentials, effects predicted to be anti-inflammatory and analgesic, respectively. Among the top dual hits are witch hazel and fireweed; polymodal modulation of multiple K+ channel types by hydrolysable tannins contributes to their dual anti-inflammatory, analgesic actions. In silico docking and mutagenesis data suggest pore-proximal extracellular linker sequence divergence underlies opposite effects of hydrolysable tannins on different Kv1 isoforms. The findings provide molecular insights into the enduring, widespread medicinal use of witch hazel and fireweed and demonstrate a screening strategy for discovering dual anti-inflammatory, analgesic small molecules.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Ryan F Yoshimura
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Andriy V Yeromin
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Derk Hogenkamp
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Jennifer van der Horst
- Department of Biomedical Sciences, Vascular Biology Group, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Angel Zavala
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Sonia Chinedu
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Grey Arena
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, USA
| | - Emma Lasky
- Redwood Creek Vegetation Team, National Park Service, Sausalito, CA, USA
| | - Mark Fisher
- Philip L. Boyd Deep Canyon Desert Research Center, University of California Natural Reserve System, Indian Wells, CA, USA
| | - Christopher R Tracy
- Philip L. Boyd Deep Canyon Desert Research Center, University of California Natural Reserve System, Indian Wells, CA, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Thomas A Jepps
- Department of Biomedical Sciences, Vascular Biology Group, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Michael D Cahalan
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
3
|
Xie AX, Iguchi N, Malykhina AP. Long-term follow-up of TREK-1 KO mice reveals the development of bladder hypertrophy and impaired bladder smooth muscle contractility with age. Am J Physiol Renal Physiol 2024; 326:F957-F970. [PMID: 38601986 PMCID: PMC11386977 DOI: 10.1152/ajprenal.00382.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Stretch-activated two-pore domain K+ (K2P) channels play important roles in many visceral organs, including the urinary bladder. The TWIK-related K+ channel TREK-1 is the predominantly expressed K2P channel in the urinary bladder of humans and rodents. Downregulation of TREK-1 channels was observed in the urinary bladder of patients with detrusor overactivity, suggesting their involvement in the pathogenesis of voiding dysfunction. This study aimed to characterize the long-term effects of TREK-1 on bladder function with global and smooth muscle-specific TREK-1 knockout (KO) mice. Bladder morphology, bladder smooth muscle (BSM) contractility, and voiding patterns were evaluated up to 12 mo of age. Both sexes were included in this study to probe the potential sex differences. Smooth muscle-specific TREK-1 KO mice were used to distinguish the effects of TREK-1 downregulation in BSM from the neural pathways involved in the control of bladder contraction and relaxation. TREK-1 KO mice developed enlarged urinary bladders (by 60.0% for males and by 45.1% for females at 6 mo; P < 0.001 compared with the age-matched control group) and had a significantly increased bladder capacity (by 137.7% at 12 mo; P < 0.0001) and compliance (by 73.4% at 12 mo; P < 0.0001). Bladder strips isolated from TREK-1 KO mice exhibited decreased contractility (peak force after KCl at 6 mo was 1.6 ± 0.7 N/g compared with 3.4 ± 2.0 N/g in the control group; P = 0.0005). The lack of TREK-1 channels exclusively in BSM did not replicate the bladder phenotype observed in TREK-1 KO mice, suggesting a strong neurogenic origin of TREK-1-related bladder dysfunction.NEW & NOTEWORTHY This study compared voiding function and bladder phenotypes in global and smooth muscle-specific TREK-1 KO mice. We found significant age-related changes in bladder contractility, suggesting that the lack of TREK-1 channel activity might contribute to age-related changes in bladder smooth muscle physiology.
Collapse
Affiliation(s)
- Alison Xiaoqiao Xie
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| | - Nao Iguchi
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| | - Anna P Malykhina
- Division of Urology, Department of SurgeryUniversity of Colorado Anschutz Medical CampusAuroraColoradoUnited States
| |
Collapse
|
4
|
Schroeter CB, Nelke C, Schewe M, Spohler L, Herrmann AM, Müntefering T, Huntemann N, Kuzikov M, Gribbon P, Albrecht S, Bock S, Hundehege P, Neelsen LC, Baukrowitz T, Seebohm G, Wünsch B, Bittner S, Ruck T, Budde T, Meuth SG. Validation of TREK1 ion channel activators as an immunomodulatory and neuroprotective strategy in neuroinflammation. Biol Chem 2023; 404:355-375. [PMID: 36774650 DOI: 10.1515/hsz-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/16/2023] [Indexed: 02/13/2023]
Abstract
Modulation of two-pore domain potassium (K2P) channels has emerged as a novel field of therapeutic strategies as they may regulate immune cell activation and metabolism, inflammatory signals, or barrier integrity. One of these ion channels is the TWIK-related potassium channel 1 (TREK1). In the current study, we report the identification and validation of new TREK1 activators. Firstly, we used a modified potassium ion channel assay to perform high-throughput-screening of new TREK1 activators. Dose-response studies helped to identify compounds with a high separation between effectiveness and toxicity. Inside-out patch-clamp measurements of Xenopus laevis oocytes expressing TREK1 were used for further validation of these activators regarding specificity and activity. These approaches yielded three substances, E1, B3 and A2 that robustly activate TREK1. Functionally, we demonstrated that these compounds reduce levels of adhesion molecules on primary human brain and muscle endothelial cells without affecting cell viability. Finally, we studied compound A2 via voltage-clamp recordings as this activator displayed the strongest effect on adhesion molecules. Interestingly, A2 lacked TREK1 activation in the tested neuronal cell type. Taken together, this study provides data on novel TREK1 activators that might be employed to pharmacologically modulate TREK1 activity.
Collapse
Affiliation(s)
- Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Marcus Schewe
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Lucas Spohler
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Alexander M Herrmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Thomas Müntefering
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune mediated diseases (CIMD), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune mediated diseases (CIMD), Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Sarah Albrecht
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Stefanie Bock
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Petra Hundehege
- Department of Neurology with Institute for Translational Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Lea Christine Neelsen
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Thomas Baukrowitz
- Institute of Physiology, Christian-Albrechts University Kiel, Hermann-Rodewald-Straße 5, 24118 Kiel, Germany
| | - Guiscard Seebohm
- IfGH-Cellular Electrophysiology, Department of Cardiology and Angiology, University Hospital of Münster, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center Mainz, Langenbeckstraße 1, D-55131 Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Robert-Koch-Straße 27A, D-48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
5
|
Zyrianova T, Lopez B, Zou K, Gu C, Pham D, Talapaneni S, Waters CM, Olcese R, Schwingshackl A. Activation of TREK-1 ( K2P2.1) potassium channels protects against influenza A-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2023; 324:L64-L75. [PMID: 36410022 PMCID: PMC9829483 DOI: 10.1152/ajplung.00116.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/05/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Influenza-A virus (IAV) infects yearly an estimated one billion people worldwide, resulting in 300,000-650,000 deaths. Preventive vaccination programs and antiviral medications represent the mainstay of therapy, but with unacceptably high morbidity and mortality rates, new targeted therapeutic approaches are urgently needed. Since inflammatory processes are commonly associated with measurable changes in the cell membrane potential (Em), we investigated whether Em hyperpolarization via TREK-1 (K2P2.1) K+ channel activation can protect against influenza-A virus (IAV)-induced pneumonia. We infected mice with IAV, which after 5 days caused 10-15% weight loss and a decrease in spontaneous activity, representing a clinically relevant infection. We then started a 3-day intratracheal treatment course with the novel TREK-1 activating compounds BL1249 or ML335. We confirmed TREK-1 activation with both compounds in untreated and IAV-infected primary human alveolar epithelial cells (HAECs) using high-throughput fluorescent imaging plate reader (FLIPR) assays. In mice, TREK-1 activation with BL1249 and ML335 counteracted IAV-induced histological lung injury and decrease in lung compliance and improved BAL fluid total protein levels, cell counts, and inflammatory IL-6, IP-10/CXCL-10, MIP-1α, and TNF-α levels. To determine whether these anti-inflammatory effects were mediated by activation of alveolar epithelial TREK-1 channels, we studied the effects of BL1249 and ML335 in IAV-infected HAEC, and found that TREK-1 activation decreased IAV-induced inflammatory IL-6, IP-10/CXCL10, and CCL-2 secretion. Dissection of TREK-1 downstream signaling pathways and construction of protein-protein interaction (PPI) networks revealed NF-κB1 and retinoic acid-inducible gene-1 (RIG-1) cascades as the most likely targets for TREK-1 protection. Therefore, TREK-1 activation may represent a novel therapeutic approach against IAV-induced lung injury.
Collapse
Affiliation(s)
- Tatiana Zyrianova
- Department of Pediatrics, University of California, Los Angeles, California
| | - Benjamin Lopez
- Department of Pediatrics, University of California, Los Angeles, California
| | - Kathlyn Zou
- Department of Pediatrics, University of California, Los Angeles, California
| | - Charles Gu
- Department of Pediatrics, University of California, Los Angeles, California
| | - Dayna Pham
- Department of Pediatrics, University of California, Los Angeles, California
| | | | | | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine, University of California, Los Angeles, California
- Department of Physiology, University of California, Los Angeles, California
| | | |
Collapse
|
6
|
Choveau FS, Ben Soussia I, Bichet D, Franck CC, Feliciangeli S, Lesage F. Convergence of Multiple Stimuli to a Single Gate in TREK1 and TRAAK Potassium Channels. Front Pharmacol 2021; 12:755826. [PMID: 34658895 PMCID: PMC8514629 DOI: 10.3389/fphar.2021.755826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022] Open
Abstract
Inhibitory potassium channels of the TREK1/TRAAK family are integrators of multiple stimuli, including temperature, membrane stretch, polyunsaturated fatty acids and pH. How these signals affect the gating of these channels is the subject of intense research. We have previously identified a cytoplasmic domain, pCt, which plays a major role in controlling channel activity. Here, we use pharmacology to show that the effects of pCt, arachidonic acid, and extracellular pH converge to the same gate within the channel. Using a state-dependent inhibitor, fluoxetine, as well as natural and synthetic openers, we provide further evidence that the “up” and “down” conformations identified by crystallography do not correspond to open and closed states of these channels.
Collapse
Affiliation(s)
- Frank S Choveau
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Ismail Ben Soussia
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Delphine Bichet
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Chatelain C Franck
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Sylvain Feliciangeli
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| | - Florian Lesage
- Université Côte D'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, LabEx ICST, Valbonne, France
| |
Collapse
|
7
|
Wang Y, Fu Z, Ma Z, Li N, Shang H. Bepridil, a class IV antiarrhythmic agent, can block the TREK-1 potassium channel. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1123. [PMID: 34430564 PMCID: PMC8350656 DOI: 10.21037/atm-20-7971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/17/2021] [Indexed: 11/24/2022]
Abstract
Background The TWIK-related potassium channel (TREK-1) can be regulated by different stimuli. However, it is not clear whether some antiarrhythmics affect the activity of TREK-1. In the present study, the effect of bepridil on the TREK-1 currents is investigated. Methods In a TREK-1 stably-expressed HEK-293 cell line (HEK-TREK-1), U251MG cells, and isolated mouse ventricular myocytes, the TREK-1 current and action potentials were recorded by the patch-clamp technique. The standard voltage protocol was a 200 ms constant potential at 20 mV, followed bya 500 ms ramp from –90 to +20 mV (HEK-TREK-1) or +80 mV (U251MG cells and myocytes) every 10 s. The currents at +20 mV or +80 mV were used for analysis. The docking study of bepridil’s binding model in the TREK-1 channel was performed using the Swissdock web service. Results In HEK-TREK-1 cells, BL1249 induced a significantly large outwardly rectifying current with similar baseline TREK-1 current characteristic, with a reversal potential (−70 mV). The concentration of half-maximal activation (EC50) of BL1249 was 3.45 µM. However, bepridil decreased the baseline TREK-1 currents, with a concentration of half-maximal inhibition (IC50) 0.59 µM and a Hill coefficient of 1.1. Also, bepridil inhibited BL1249-activated TREK-1 currents, with an IC50 4.08 µM and a Hill coefficient of 3.22. The outside-out patch-clamp confirmed bepridil inhibited BL1249-activated TREK-1 currents. In U251MG cells and myocytes, BL1249 activated outwardly rectifying endogenous TREK-1 currents, which could be inhibited by bepridil. BL1249 (10 µM) could decrease the peak value and reduce the duration of the action potential. Bepridil (10 µM) prolonged the duration of action potential of myocytes. The docking study revealed that bepridil might affect the K+ pore domain and the M4 modulator pocket. Conclusions Bepridil may be a blocker for the TREK-1K+channel at a clinically therapeutic concentration, providing a new mechanism of TREK-1 regulation and bepridil's antiarrhythmic effect.
Collapse
Affiliation(s)
- Ying Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China
| | - Zhijie Fu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China.,Department of Otorhinolaryngology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Zhiyong Ma
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China
| | - Na Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China
| | - Hong Shang
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
8
|
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
|
9
|
García G, Martínez-Rojas VA, Murbartián J. TREK-1 potassium channels participate in acute and long-lasting nociceptive hypersensitivity induced by formalin in rats. Behav Brain Res 2021; 413:113446. [PMID: 34224765 DOI: 10.1016/j.bbr.2021.113446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/18/2021] [Accepted: 07/01/2021] [Indexed: 01/06/2023]
Abstract
TREK-1 channels are expressed in small nociceptive dorsal root ganglion (DRG) neurons where they participate in acute inflammatory and neuropathic pain. However, the role of TREK-1 in persistent pain is not well understood. The aim of this study was to investigate the local peripheral and spinal participation of TREK-1 in formalin-induced acute and long-lasting nociceptive hypersensitivity. Local peripheral or intrathecal pre-treatment with spadin, selective blocker of TREK-1, increased acute flinching behavior and secondary mechanical allodynia and hyperalgesia behavior observed 6 days after formalin injection. Local peripheral or intrathecal pre-treatment with BL-1249, selective opener of TREK-1, decreased long-lasting secondary mechanical allodynia and hyperalgesia induced by formalin. Pre-treatment with BL-1249 prevented the pro-nociceptive effect of spadin on acute nociception and long-lasting mechanical allodynia and hyperalgesia in rats. Pre-treatment with two recombinant channels that produce a high TREK-1 current, S300A and S333A (non-phosphorylated state of TREK-1), reduced formalin-induced acute pain and long-lasting mechanical allodynia and hyperalgesia. Besides, post-treatment with S300A, S333A or BL-1249 reversed long-lasting mechanical allodynia and hyperalgesia induced by formalin. Formalin increased TREK-1 expression at 1 and 6 days in DRG and dorsal spinal cord in rats, whereas that it increased c-fos expression at the DRG. Intrathecal repeated transfection of rats with S300A and S333A or injection with BL-1249 reduced formalin-induced enhanced c-fos expression. Data suggest that TREK-1 activity at peripheral and spinal sites reduces neuronal excitability in the process of acute and long-lasting nociception induced by formalin in rats.
Collapse
Affiliation(s)
- Guadalupe García
- Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico.
| | | | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, Sede Sur, Mexico City, Mexico.
| |
Collapse
|
10
|
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
|
11
|
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: 47] [Impact Index Per Article: 11.8] [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
|
12
|
Huang L, Xu G, Jiang R, Luo Y, Zuo Y, Liu J. Development of Non-opioid Analgesics Targeting Two-pore Domain Potassium Channels. Curr Neuropharmacol 2021; 20:16-26. [PMID: 33827408 PMCID: PMC9199554 DOI: 10.2174/1570159x19666210407152528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Two-pore domain potassium (K2P) channels are a diverse family of potassium channels. K2P channels generate background leak potassium currents to regulate cellular excitability and are thereby involved in a wide range of neurological disorders. K2P channels are modulated by a variety of physicochemical factors such as mechanical stretch, temperature, and pH. In the the peripheral nervous system (PNS), K2P channels are widely expressed in nociceptive neurons and play a critical roles in pain perception. In this review, we summarize the recent advances in the pharmacological properties of K2P channels, with a focus on the exogenous small-molecule activators targeting K2P channels. We emphasize the subtype-selectivity, cellular and in vivo pharmacological properties of all the reported small-molecule activators. The key underlying analgesic mechanisms mediated by K2P are also summarized based on the data in the literature from studies using small-molecule activators and genetic knock-out animals. We discuss advantages and limitations of the translational perspectives of K2P in pain medicine and provide outstanding questions for future studies in the end.
Collapse
Affiliation(s)
- Lu Huang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Guangyin Xu
- Department of Physiology and Neurobiology, Institute of Neuroscience, Medical College of Soochow University, Suzhou, 215123, Jiangsu. China
| | - Ruotian Jiang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yuncheng Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Yunxia Zuo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan. China
| |
Collapse
|
13
|
|
14
|
McCoull D, Ococks E, Large JM, Tickle DC, Mathie A, Jerman J, Wright PD. A "Target Class" Screen to Identify Activators of Two-Pore Domain Potassium (K2P) Channels. SLAS DISCOVERY 2020; 26:428-438. [PMID: 33375888 PMCID: PMC7900820 DOI: 10.1177/2472555220976126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Two-pore domain potassium (K2P) channels carry background (or leak) potassium
current and play a key role in regulating resting membrane potential and
cellular excitability. Accumulating evidence points to a role for K2Ps in human
pathophysiologies, most notably in pain and migraine, making them attractive
targets for therapeutic intervention. However, there remains a lack of selective
pharmacological tools. The aim of this work was to apply a “target class”
approach to investigate the K2P superfamily and identify novel activators across
all the described subclasses of K2P channels. Target class drug discovery allows
for the leveraging of accumulated knowledge and maximizing synergies across a
family of targets and serves as an additional approach to standard target-based
screening. A common assay platform using baculovirus (BacMam) to transiently
express K2P channels in mammalian cells and a thallium flux assay to determine
channel activity was developed, allowing the simultaneous screening of multiple
targets. Importantly, this system, by allowing precise titration of channel
function, allows optimization to facilitate the identification of activators. A
representative set of channels (THIK-1, TWIK-1, TREK-2, TASK-3, and TASK-2) were
screened against a library of Food and Drug Administration (FDA)-approved
compounds and the LifeArc Index Set. Activators were then analyzed in
concentration–response format across all channels to assess selectivity. Using
the target class approach to investigate the K2P channels has enabled us to
determine which of the K2Ps are amenable to small-molecule activation, de-risk
multiple channels from a technical point of view, and identify a diverse range
of previously undescribed pharmacology.
Collapse
Affiliation(s)
| | | | | | | | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent, UK
| | | | | |
Collapse
|
15
|
K 2P2.1 (TREK-1) potassium channel activation protects against hyperoxia-induced lung injury. Sci Rep 2020; 10:22011. [PMID: 33319831 PMCID: PMC7738539 DOI: 10.1038/s41598-020-78886-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
No targeted therapies exist to counteract Hyperoxia (HO)-induced Acute Lung Injury (HALI). We previously found that HO downregulates alveolar K2P2.1 (TREK-1) K+ channels, which results in worsening lung injury. This decrease in TREK-1 levels leaves a subset of channels amendable to pharmacological intervention. Therefore, we hypothesized that TREK-1 activation protects against HALI. We treated HO-exposed mice and primary alveolar epithelial cells (AECs) with the novel TREK-1 activators ML335 and BL1249, and quantified physiological, histological, and biochemical lung injury markers. We determined the effects of these drugs on epithelial TREK-1 currents, plasma membrane potential (Em), and intracellular Ca2+ (iCa) concentrations using fluorometric assays, and blocked voltage-gated Ca2+ channels (CaV) as a downstream mechanism of cytokine secretion. Once-daily, intra-tracheal injections of HO-exposed mice with ML335 or BL1249 improved lung compliance, histological lung injury scores, broncho-alveolar lavage protein levels and cell counts, and IL-6 and IP-10 concentrations. TREK-1 activation also decreased IL-6, IP-10, and CCL-2 secretion from primary AECs. Mechanistically, ML335 and BL1249 induced TREK-1 currents in AECs, counteracted HO-induced cell depolarization, and lowered iCa2+ concentrations. In addition, CCL-2 secretion was decreased after L-type CaV inhibition. Therefore, Em stabilization with TREK-1 activators may represent a novel approach to counteract HALI.
Collapse
|
16
|
García G, Méndez-Reséndiz KA, Oviedo N, Murbartián J. PKC- and PKA-dependent phosphorylation modulates TREK-1 function in naïve and neuropathic rats. J Neurochem 2020; 157:2039-2054. [PMID: 33006141 DOI: 10.1111/jnc.15204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
PKC and PKA phosphorylation inhibit TREK-1 channels downstream of Gs protein-coupled receptor activation in vitro. However, the role of phosphorylation of TREK-1 in neuropathic pain is unknown. The purpose of this study was to investigate whether altered TREK-1 channel function by PKA and PKC modulators contributes to antiallodynia in neuropathic rats. Furthermore, we investigated if the in vitro described sites for PKC and PKA phosphorylation (S300 and S333, respectively) participate in the modulation of TREK-1 in naïve and neuropathic rats. L5/L6 spinal nerve ligation (SNL) induced tactile allodynia. Intrathecal injection of BL-1249 (TREK-1 activator) reversed nerve injury-induced tactile allodynia, whereas spadin (TREK-1 blocker) produced tactile allodynia in naïve rats and reversed the antiallodynic effect induced by BL-1249 in neuropathic rats. Intrathecal administration of rottlerin or Rp-cAMPs (PKC and PKA inhibitors, respectively) enhanced the antiallodynia observed with BL-1249 in neuropathic rats. In contrast, pretreatment with PdBu or forskolin (PKC and PKA activators, respectively) reduced the BL-1249-induced antiallodynia. Intrathecal injection of two high-activity TREK-1 recombinant channels, using a in vivo transfection method with lipofectamine, with mutations at PKC/PKA phosphosites (S300A and S333A) reversed tactile allodynia in neuropathic rats, with no effect in naïve rats. In contrast, transfection of two low-activity TREK-1 recombinant channels with phosphomimetic mutations at those sites (S300D and S333D) produced tactile allodynia in naïve rats and interfered with antiallodynic effects of rottlerin/BL-1249 or Rp-cAMPs/BL-1249. Data suggest that TREK-1 channel activity can be dynamically tuned in vivo by PKC/PKA to provoke allodynia and modulate its antiallodynic role in neuropathic pain.
Collapse
Affiliation(s)
- Guadalupe García
- Departamento de Farmacobiología, Cinvestav, Sede Sur., Mexico City, Mexico
| | | | - Norma Oviedo
- Unidad de Investigación Médica en Inmunología e Infectología, Centro Médico Nacional, La Raza, Instituto Mexicano del Seguro Social., Mexico City, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, Sede Sur., Mexico City, Mexico
| |
Collapse
|
17
|
Malysz J, Petkov GV. Urinary bladder smooth muscle ion channels: expression, function, and regulation in health and disease. Am J Physiol Renal Physiol 2020; 319:F257-F283. [PMID: 32628539 PMCID: PMC7473901 DOI: 10.1152/ajprenal.00048.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/21/2020] [Accepted: 06/28/2020] [Indexed: 12/17/2022] Open
Abstract
Urinary bladder smooth muscle (UBSM), also known as detrusor smooth muscle, forms the bladder wall and ultimately determines the two main attributes of the organ: urine storage and voiding. The two functions are facilitated by UBSM relaxation and contraction, respectively, which depend on UBSM excitability shaped by multiple ion channels. In this review, we summarize the current understanding of key ion channels establishing and regulating UBSM excitability and contractility. They include excitation-enhancing voltage-gated Ca2+ (Cav) and transient receptor potential channels, excitation-reducing K+ channels, and still poorly understood Cl- channels. Dynamic interplay among UBSM ion channels determines the overall level of Cav channel activity. The net Ca2+ influx via Cav channels increases global intracellular Ca2+ concentration, which subsequently triggers UBSM contractility. Here, for each ion channel type, we describe UBSM tissue/cell expression (mRNA and protein) profiles and their role in regulating excitability and contractility of UBSM in various animal species, including the mouse, rat, and guinea pig, and, most importantly, humans. The currently available data reveal certain interspecies differences, which complicate the translational value of published animal research results to humans. This review highlights recent developments, findings on genetic knockout models, pharmacological data, reports on UBSM ion channel dysfunction in animal bladder disease models, and the very limited human studies currently available. Among all gaps in present-day knowledge, the unknowns on expression and functional roles for ion channels determined directly in human UBSM tissues and cells under both normal and disease conditions remain key hurdles in the field.
Collapse
Affiliation(s)
- John Malysz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Georgi V Petkov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Urology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
18
|
Mathie A, Veale EL, Cunningham KP, Holden RG, Wright PD. Two-Pore Domain Potassium Channels as Drug Targets: Anesthesia and Beyond. Annu Rev Pharmacol Toxicol 2020; 61:401-420. [PMID: 32679007 DOI: 10.1146/annurev-pharmtox-030920-111536] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two-pore domain potassium (K2P) channels stabilize the resting membrane potential of both excitable and nonexcitable cells and, as such, are important regulators of cell activity. There are many conditions where pharmacological regulation of K2P channel activity would be of therapeutic benefit, including, but not limited to, atrial fibrillation, respiratory depression, pulmonary hypertension, neuropathic pain, migraine, depression, and some forms of cancer. Up until now, few if any selective pharmacological regulators of K2P channels have been available. However, recent publications of solved structures with small-molecule activators and inhibitors bound to TREK-1, TREK-2, and TASK-1 K2P channels have given insight into the pharmacophore requirements for compound binding to these sites. Together with the increasing availability of a number of novel, active, small-molecule compounds from K2P channel screening programs, these advances have opened up the possibility of rational activator and inhibitor design to selectively target K2P channels.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Emma L Veale
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | - Kevin P Cunningham
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - Robyn G Holden
- Medway School of Pharmacy, University of Greenwich and University of Kent, Kent ME4 4TB, United Kingdom;
| | | |
Collapse
|
19
|
Rivas-Ramírez P, Reboreda A, Rueda-Ruzafa L, Herrera-Pérez S, Lamas JA. PIP 2 Mediated Inhibition of TREK Potassium Currents by Bradykinin in Mouse Sympathetic Neurons. Int J Mol Sci 2020; 21:ijms21020389. [PMID: 31936257 PMCID: PMC7014146 DOI: 10.3390/ijms21020389] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
Bradykinin (BK), a hormone inducing pain and inflammation, is known to inhibit potassium M-currents (IM) and to increase the excitability of the superior cervical ganglion (SCG) neurons by activating the Ca2+-calmodulin pathway. M-current is also reduced by muscarinic agonists through the depletion of membrane phosphatidylinositol 4,5-biphosphate (PIP2). Similarly, the activation of muscarinic receptors inhibits the current through two-pore domain potassium channels (K2P) of the “Tandem of pore-domains in a Weakly Inward rectifying K+ channel (TWIK)-related channels” (TREK) subfamily by reducing PIP2 in mouse SCG neurons (mSCG). The aim of this work was to test and characterize the modulation of TREK channels by bradykinin. We used the perforated-patch technique to investigate riluzole (RIL) activated currents in voltage- and current-clamp experiments. RIL is a drug used in the palliative treatment of amyotrophic lateral sclerosis and, in addition to blocking voltage-dependent sodium channels, it also selectively activates the K2P channels of the TREK subfamily. A cell-attached patch-clamp was also used to investigate TREK-2 single channel currents. We report here that BK reduces spike frequency adaptation (SFA), inhibits the riluzole-activated current (IRIL), which flows mainly through TREK-2 channels, by about 45%, and reduces the open probability of identified single TREK-2 channels in cultured mSCG cells. The effect of BK on IRIL was precluded by the bradykinin receptor (B2R) antagonist HOE-140 (d-Arg-[Hyp3, Thi5, d-Tic7, Oic8]BK) but also by diC8PIP2 which prevents PIP2 depletion when phospholipase C (PLC) is activated. On the contrary, antagonizing inositol triphosphate receptors (IP3R) using 2-aminoethoxydiphenylborane (2-APB) or inhibiting protein kinase C (PKC) with bisindolylmaleimide did not affect the inhibition of IRIL by BK. In conclusion, bradykinin inhibits TREK-2 channels through the activation of B2Rs resulting in PIP2 depletion, much like we have demonstrated for muscarinic agonists. This mechanism implies that TREK channels must be relevant for the capture of information about pain and visceral inflammation.
Collapse
|
20
|
Al-Moubarak E, Veale EL, Mathie A. Pharmacologically reversible, loss of function mutations in the TM2 and TM4 inner pore helices of TREK-1 K2P channels. Sci Rep 2019; 9:12394. [PMID: 31455781 PMCID: PMC6712037 DOI: 10.1038/s41598-019-48855-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/09/2019] [Indexed: 01/21/2023] Open
Abstract
A better understanding of the gating of TREK two pore domain potassium (K2P) channels and their activation by compounds such as the negatively charged activator, flufenamic acid (FFA) is critical in the search for more potent and selective activators of these channels. Currents through wild-type and mutated human K2P channels expressed in tsA201 cells were measured using whole-cell patch-clamp recordings in the presence and absence of FFA. Mutation of the TM2.6 residue of TREK-1 to a phenylalanine (G171F) and a similar mutation of TM4.6 (A286F) substantially reduced current through TREK-1 channels. In complementary experiments, replacing the natural F residues at the equivalent position in TRESK channels, significantly enhanced current. Known, gain of function mutations of TREK-1 (G137I, Y284A) recovered current through these mutated channels. This reduction in current could be also be reversed pharmacologically, by FFA. However, an appropriate length MTS (MethaneThioSulfonate) cross-linking reagent (MTS14) restricted the activation of TREK-1_A286C channels by repeated application of FFA. This suggests that the cross-linker stabilises the channel in a conformation which blunts FFA activation. Pharmacologically reversible mutations of TREK channels will help to clarify the importance of these channels in pathophysiological conditions such as pain and depression.
Collapse
Affiliation(s)
- Ehab Al-Moubarak
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Kent, ME4 4TB, UK.
| |
Collapse
|
21
|
Towards a TREK-1/2 (TWIK-Related K+ Channel 1 and 2) dual activator tool compound: Multi-dimensional optimization of BL-1249. Bioorg Med Chem Lett 2019; 29:1601-1604. [DOI: 10.1016/j.bmcl.2019.04.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 11/20/2022]
|
22
|
Djillani A, Mazella J, Heurteaux C, Borsotto M. Role of TREK-1 in Health and Disease, Focus on the Central Nervous System. Front Pharmacol 2019; 10:379. [PMID: 31031627 PMCID: PMC6470294 DOI: 10.3389/fphar.2019.00379] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/26/2019] [Indexed: 01/22/2023] Open
Abstract
TREK-1 is the most studied background K2P channel. Its main role is to control cell excitability and maintain the membrane potential below the threshold of depolarization. TREK-1 is multi-regulated by a variety of physical and chemical stimuli which makes it a very promising and challenging target in the treatment of several pathologies. It is mainly expressed in the brain but also in heart, smooth muscle cells, endocrine pancreas, and prostate. In the nervous system, TREK-1 is involved in many physiological and pathological processes such as depression, neuroprotection, pain, and anesthesia. These properties explain why many laboratories and pharmaceutical companies have been focusing their research on screening and developing highly efficient modulators of TREK-1 channels. In this review, we summarize the different roles of TREK-1 that have been investigated so far in attempt to characterize pharmacological tools and new molecules to modulate cellular functions controlled by TREK-1.
Collapse
Affiliation(s)
- Alaeddine Djillani
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Jean Mazella
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Catherine Heurteaux
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| | - Marc Borsotto
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France
| |
Collapse
|
23
|
Tarasov MV, Kotova PD, Bystrova MF, Kabanova NV, Sysoeva VY, Kolesnikov SS. Arachidonic acid hyperpolarizes mesenchymal stromal cells from the human adipose tissue by stimulating TREK1 K + channels. Channels (Austin) 2019; 13:36-47. [PMID: 30661462 PMCID: PMC6380217 DOI: 10.1080/19336950.2019.1565251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The current knowledge of electrogenesis in mesenchymal stromal cells (MSCs) remains scarce. Earlier, we demonstrated that in MSCs from the human adipose tissue, transduction of certain agonists involved the phosphoinositide cascade. Its pivotal effector PLC generates DAG that can regulate ion channels directly or via its derivatives, including arachidonic acid (AA). Here we showed that AA strongly hyperpolarized MSCs by stimulating instantly activating, outwardly rectifying TEA-insensitive K+ channels. Among AA-regulated K+ channels, K2P channels from the TREK subfamily appeared to be an appropriate target. The expression of K2P channels in MSCs was verified by RT-PCR, which revealed TWIK-1, TREK-1, and TASK-5 transcripts. The TREK-1 inhibitor spadin antagonized the electrogenic action of AA, which was simulated by the channel activator BL 1249. This functional evidence suggested that TREK-1 channels mediated AA-dependent hyperpolarization of MSCs. Being mostly silent at rest, TREK-1 negligibly contributed to the “background” K+ current. The dramatic stimulation of TREK-1 channels by AA indicates their involvement in AA-dependent signaling in MSCs.
Collapse
Affiliation(s)
- Michail V Tarasov
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Polina D Kotova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Marina F Bystrova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Natalia V Kabanova
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| | - Veronika Yu Sysoeva
- b Department of Biochemistry and Molecular Medicine, Faculty of Basic Medicine , Lomonosov Moscow State University , Moscow , Russia
| | - Stanislav S Kolesnikov
- a Department of Molecular Cell Physiology, Institute of Cell Biophysics , Russian Academy of Sciences , Pushchino , Moscow Region , Russia
| |
Collapse
|
24
|
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
|
25
|
Fernández-Fernández D, Cadaveira-Mosquera A, Rueda-Ruzafa L, Herrera-Pérez S, Veale EL, Reboreda A, Mathie A, Lamas JA. Activation of TREK currents by riluzole in three subgroups of cultured mouse nodose ganglion neurons. PLoS One 2018; 13:e0199282. [PMID: 29928032 PMCID: PMC6013220 DOI: 10.1371/journal.pone.0199282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/05/2018] [Indexed: 01/12/2023] Open
Abstract
Two-pore domain potassium channels (K2P) constitute major candidates for the regulation of background potassium currents in mammalian cells. Channels of the TREK subfamily are also well positioned to play an important role in sensory transduction due to their sensitivity to a large number of physiological and physical stimuli (pH, mechanical, temperature). Following our previous report describing the molecular expression of different K2P channels in the vagal sensory system, here we confirm that TREK channels are functionally expressed in neurons from the mouse nodose ganglion (mNG). Neurons were subdivided into three groups (A, Ah and C) based on their response to tetrodotoxin and capsaicin. Application of the TREK subfamily activator riluzole to isolated mNG neurons evoked a concentration-dependent outward current in the majority of cells from all the three subtypes studied. Riluzole increased membrane conductance and hyperpolarized the membrane potential by approximately 10 mV when applied to resting neurons. The resting potential was similar in all three groups, but C cells were clearly less excitable and showed smaller hyperpolarization-activated currents at -100 mV and smaller sustained currents at -30 mV. Our results indicate that the TREK subfamily of K2P channels might play an important role in the maintenance of the resting membrane potential in sensory neurons of the autonomic nervous system, suggesting its participation in the modulation of vagal reflexes.
Collapse
Affiliation(s)
- Diego Fernández-Fernández
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
- * E-mail: (DFF); (JAL)
| | - Alba Cadaveira-Mosquera
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
| | - Lola Rueda-Ruzafa
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
| | - Salvador Herrera-Pérez
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
| | - Emma L. Veale
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent, United Kingdom
| | - Antonio Reboreda
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent, United Kingdom
| | - J. Antonio Lamas
- Department of Functional Biology and Health Sciences, Faculty of Biology–CINBIO, University of Vigo, Vigo, Galicia, Spain
- * E-mail: (DFF); (JAL)
| |
Collapse
|
26
|
Loucif AJC, Saintot P, Liu J, Antonio BM, Zellmer SG, Yoger K, Veale EL, Wilbrey A, Omoto K, Cao L, Gutteridge A, Castle NA, Stevens EB, Mathie A. GI-530159, a novel, selective, mechanosensitive two-pore-domain potassium (K 2P ) channel opener, reduces rat dorsal root ganglion neuron excitability. Br J Pharmacol 2018; 175:2272-2283. [PMID: 29150838 PMCID: PMC5980259 DOI: 10.1111/bph.14098] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE TREK two-pore-domain potassium (K2P ) channels play a critical role in regulating the excitability of somatosensory nociceptive neurons and are important mediators of pain perception. An understanding of the roles of TREK channels in pain perception and, indeed, in other pathophysiological conditions, has been severely hampered by the lack of potent and/or selective activators and inhibitors. In this study, we describe a new, selective opener of TREK channels, GI-530159. EXPERIMENTAL APPROACH The effect of GI-530159 on TREK channels was demonstrated using 86 Rb efflux assays, whole-cell and single-channel patch-clamp recordings from recombinant TREK channels. The expression of K2P 2.1 (TREK1), K2P 10.1 (TREK2) and K2P 4.1 (TRAAK) channels was determined using transcriptome analysis from single dorsal root ganglion (DRG) cells. Current-clamp recordings from cultured rat DRG neurons were used to measure the effect of GI-530159 on neuronal excitability. KEY RESULTS For recombinant human TREK1 channels, GI-530159 had similar low EC50 values in Rb efflux experiments and electrophysiological recordings. It activated TREK2 channels, but it had no detectable action on TRAAK channels nor any significant effect on other K channels tested. Current-clamp recordings from cultured rat DRG neurones showed that application of GI-530159 at 1 μM resulted in a significant reduction in firing frequency and a small hyperpolarization of resting membrane potential. CONCLUSIONS AND IMPLICATIONS This study provides pharmacological evidence for the presence of mechanosensitive TREK K2P channels in sensory neurones and suggests that development of selective K2P channel openers like GI-530159 could aid in the development of novel analgesic agents. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emma L Veale
- Medway School of PharmacyUniversity of KentChatham MaritimeKentUK
| | | | | | | | | | | | | | - Alistair Mathie
- Medway School of PharmacyUniversity of KentChatham MaritimeKentUK
| |
Collapse
|
27
|
Ma R, Seifi M, Papanikolaou M, Brown JF, Swinny JD, Lewis A. TREK-1 Channel Expression in Smooth Muscle as a Target for Regulating Murine Intestinal Contractility: Therapeutic Implications for Motility Disorders. Front Physiol 2018; 9:157. [PMID: 29563879 PMCID: PMC5845753 DOI: 10.3389/fphys.2018.00157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
Gastrointestinal (GI) motility disorders such as irritable bowel syndrome (IBS) can occur when coordinated smooth muscle contractility is disrupted. Potassium (K+) channels regulate GI smooth muscle tone and are key to GI tract relaxation, but their molecular and functional phenotypes are poorly described. Here we define the expression and functional roles of mechano-gated K2P channels in mouse ileum and colon. Expression and distribution of the K2P channel family were investigated using quantitative RT-PCR (qPCR), immunohistochemistry and confocal microscopy. The contribution of mechano-gated K2P channels to mouse intestinal muscle tension was studied pharmacologically using organ bath. Multiple K2P gene transcripts were detected in mouse ileum and colon whole tissue preparations. Immunohistochemistry confirmed TREK-1 expression was smooth muscle specific in both ileum and colon, whereas TREK-2 and TRAAK channels were detected in enteric neurons but not smooth muscle. In organ bath, mechano-gated K2P channel activators (Riluzole, BL-1249, flufenamic acid, and cinnamyl 1-3,4-dihydroxy-alpha-cyanocinnamate) induced relaxation of KCl and CCh pre-contracted ileum and colon tissues and reduced the amplitude of spontaneous contractions. These data reveal the specific expression of mechano-gated K2P channels in mouse ileum and colon tissues and highlight TREK-1, a smooth muscle specific K2P channel in GI tract, as a potential therapeutic target for combating motility pathologies arising from hyper-contractility.
Collapse
Affiliation(s)
- Ruolin Ma
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Mohsen Seifi
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Maria Papanikolaou
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - James F Brown
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Jerome D Swinny
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Anthony Lewis
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
28
|
Lolicato M, Arrigoni C, Mori T, Sekioka Y, Bryant C, Clark KA, Minor DL. K 2P2.1 (TREK-1)-activator complexes reveal a cryptic selectivity filter binding site. Nature 2017; 547:364-368. [PMID: 28693035 PMCID: PMC5778891 DOI: 10.1038/nature22988] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022]
Abstract
Polymodal K2P (KCNK) thermo- and mechanosensitive TREK1 potassium channels, generate ‘leak’ currents that regulate neuronal excitability, respond to lipids, temperature, and mechanical stretch, and influence pain, temperature perception, and anesthetic responses1–3. These dimeric voltage-gated ion channel (VGIC) superfamily members have a unique topology comprising two pore forming regions per subunit4–6. Contrasting other potassium channels, K2Ps use a selectivity filter ‘C-type’ gate7–10 as the principal gating site. Despite recent advances3,11,12, K2Ps suffer from a poor pharmacologic profile limiting mechanistic and biological studies. Here, we describe a new small molecule TREK activator class that directly stimulates the C-type gate by acting as molecular wedges that restrict interdomain interface movement behind the selectivity filter. Structures of K2P2.1(TREK-1) alone with two selective K2P2.1(TREK-1) and K2P10.1(TREK-2) activators, an N-aryl-sulfonamide, ML335, and a thiophene-carboxamide, ML402, define a cryptic binding pocket unlike other ion channel small molecule binding sites and, together with functional studies, identify a cation-π interaction that controls selectivity. Together, our data unveil a previously unknown, druggable K2P site that stabilizes the C-type gate ‘leak mode’ and provide direct evidence for K2P selectivity filter gating.
Collapse
Affiliation(s)
- Marco Lolicato
- Cardiovascular Research Institute, University of California, San Francisco, California 941158-9001, USA
| | - Cristina Arrigoni
- Cardiovascular Research Institute, University of California, San Francisco, California 941158-9001, USA
| | - Takahiro Mori
- Ono Pharmaceutical Co. Ltd, Mishima-Gun, Osaka 618-8585, Japan
| | - Yoko Sekioka
- Ono Pharmaceutical Co. Ltd, Mishima-Gun, Osaka 618-8585, Japan
| | - Clifford Bryant
- Small Molecule Discovery Center, University of California, San Francisco, California 93858-2330, USA
| | - Kimberly A Clark
- Cardiovascular Research Institute, University of California, San Francisco, California 941158-9001, USA
| | - Daniel L Minor
- Cardiovascular Research Institute, University of California, San Francisco, California 941158-9001, USA.,Departments of Biochemistry and Biophysics, and Cellular and Molecular Pharmacology, University of California, San Francisco, California 941158-9001, USA.,California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 941158-9001, USA.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California 941158-9001, USA.,Molecular Biophysics and Integrated Bio-imaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| |
Collapse
|
29
|
pH-sensitive K+ channel TREK-1 is a novel target in pancreatic cancer. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1994-2003. [DOI: 10.1016/j.bbadis.2016.07.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 06/16/2016] [Accepted: 07/15/2016] [Indexed: 12/17/2022]
|
30
|
Monteillier A, Loucif A, Omoto K, Stevens EB, Lainez S, Saintot PP, Cao L, Pryde DC. Investigation of the structure activity relationship of flufenamic acid derivatives at the human TRESK channel K 2P 18.1. Bioorg Med Chem Lett 2016; 26:4919-4924. [DOI: 10.1016/j.bmcl.2016.09.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/30/2022]
|
31
|
Fukasaku M, Kimura J, Yamaguchi O. Swelling-activated and arachidonic acid-induced currents are TREK-1 in rat bladder smooth muscle cells. Fukushima J Med Sci 2016; 62:18-26. [PMID: 26911303 DOI: 10.5387/fms.2015-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Using the perforated patch voltage clamp, we investigated swelling-activated ionic channels (SACs) in rat urinary bladder smooth muscle cells. Hypo-osmotic (60%) bath solution increased a membrane current which was inhibited by the SAC inhibitor, gadolinium. The reversal potential of the hypotonicity-induced current shifted in the positive direction by increasing external K(+) concentration. The hypotonicity-induced current was inhibited by extracellular acidic pH, phorbol ester and forskolin. These pharmacological properties are identical to those of arachidonic acid-induced current present in these cells, suggesting the presence of TREK-1, a four-transmembrane two pore domain K(+) channel. Using RT-PCR we screened rat bladder smooth muscles and cerebellum for expression of TREK-1, TREK-2 and TRAAK mRNAs. Only TREK-1 mRNA was expressed in the bladder, while all three were expressed in the cerebellum. We conclude that a mechanosensitive K(+) channel is present in rat bladder myocytes, which is activated by arachidonic acid and most likely is TREK-1. This K(+) channel may have an important role in the regulation of bladder smooth muscle tone during urine storage.
Collapse
|
32
|
Sacco E, Recupero S, Bientinesi R, Palermo G, D’Agostino D, Currò D, Bassi P. Pioneering drugs for overactive bladder and detrusor overactivity: Ongoing research and future directions. World J Obstet Gynecol 2015; 4:24-39. [DOI: 10.5317/wjog.v4.i2.24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 01/31/2015] [Accepted: 04/14/2015] [Indexed: 02/05/2023] Open
Abstract
The ongoing research on pioneering drug candidates for the overactive bladder (OAB) aimed to overcome the limitations of currently licensed pharmacotherapies, such as antimuscarinics, β3-adrenergic agents, and botulinum neurotoxin, has been reviewed performing a systematic literature review and web search. The review covers the exploratory agents alternative to available medications for OAB and that may ultimately prove to be therapeutically useful in the future management of OAB patients based on preclinical and early clinical data. It emerges that many alternative pharmacological strategies have been discovered or are under investigation in disease-oriented studies. Several potential therapeutics are known for years but still find obstacles to pass the clinical stages of development, while other completely novel compounds, targeting new pharmacological targets, have been recently discovered and show potential to translate into clinical therapeutic agents for idiopathic and neurogenic OAB syndrome. The global scenario of investigational drugs for OAB gives promise for the development of innovative therapeutics that may ultimately prove effective as first, combined or second-line treatments within a realistic timescale of ten years.
Collapse
|
33
|
Mathie A, Veale EL. Two-pore domain potassium channels: potential therapeutic targets for the treatment of pain. Pflugers Arch 2014; 467:931-43. [DOI: 10.1007/s00424-014-1655-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 01/01/2023]
|
34
|
Sacco E, Bientinesi R. Innovative pharmacotherapies for women with overactive bladder: where are we now and what is in the pipeline? Int Urogynecol J 2014; 26:629-40. [PMID: 25377296 DOI: 10.1007/s00192-014-2557-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/18/2014] [Indexed: 12/12/2022]
Abstract
INTRODUCTION AND HYPOTHESIS The impressive prevalence of overactive bladder (OAB) and the relevant limitations of current treatments urge the need for novel therapeutic approaches. METHODS A systematic literature and web search was performed to identify investigational drugs that entered the early and late phases of clinical development for women with OAB symptoms. RESULTS Approved pharmacological therapies for OAB (antimuscarinics, beta-3 agonists, and botulinum toxin) are evolving with the development of alternative administration methods, combination strategies, and novel compounds, expected to improve effectiveness, bladder selectivity, and dose flexibility. A wealth of investigational compounds, developed with both public and companies' indoor nonclinical disease-oriented studies, entered the early and late stages of clinical development in the last decade. Most non-anticholinergic compounds in ongoing clinical trials target central and peripheral neurotransmitter receptors involved in neurological modulation of micturition, nonadrenergic-noncholinergic mechanisms, cyclic nucleotide metabolism, different subtypes of ion channels or peripheral receptors of prostaglandins, vanilloids, vitamin D3, and opioids. Fascinating advances are ongoing also in the field of genetic therapy. CONCLUSIONS New pharmaceutical formulations and drug combinations are expected to be available in the next decade in order to overcome the limitations of current drugs for OAB. Although proof-of-concept, patient-oriented studies yielded disappointing results for several tentative drugs, a lot of clinical research is ongoing that is expected to provide clinicians with novel therapeutic agents in the near future.
Collapse
Affiliation(s)
- Emilio Sacco
- Department of Urology, "Agostino Gemelli" Hospital, Catholic University Medical School, Rome, Italy,
| | | |
Collapse
|
35
|
Lei Q, Pan XQ, Chang S, Malkowicz SB, Guzzo TJ, Malykhina AP. Response of the human detrusor to stretch is regulated by TREK-1, a two-pore-domain (K2P) mechano-gated potassium channel. J Physiol 2014; 592:3013-30. [PMID: 24801307 DOI: 10.1113/jphysiol.2014.271718] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The mechanisms of mechanosensitivity underlying the response of the human bladder to stretch are poorly understood. Animal data suggest that stretch-activated two-pore-domain (K2P) K(+) channels play a critical role in bladder relaxation during the filling phase. The objective of this study was to characterize the expression and function of stretch-activated K2P channels in the human bladder and to clarify their physiological role in bladder mechanosensitivity. Gene and protein analysis of the K2P channels TREK-1, TREK-2 and TRAAK in the human bladder revealed that TREK-1 is the predominantly expressed member of the mechano-gated subfamily of K2P channels. Immunohistochemical labelling of bladder wall identified higher levels of expression of TREK-1 in detrusor smooth muscle cells in comparison to bladder mucosa. Functional characterization and biophysical properties of the predominantly expressed member of the K2P family, the TREK-1 channel, were evaluated by in vitro organ bath studies and the patch-clamp technique. Electrophysiological recordings from single smooth muscle cells confirmed direct activation of TREK-1 channels by mechanical stretch and negative pressure applied to the cell membrane. Inhibition of TREK-1 channels in the human detrusor significantly delayed relaxation of the stretched bladder smooth muscle strips and triggered small-amplitude spontaneous contractions. Application of negative pressure to cell-attached patches (-20 mmHg) caused a 19-fold increase in the open probability (NPo) of human TREK-1 channels. l-Methionine (1 mm), a specific TREK-1 inhibitor, dramatically decreased the NPo of TREK-1 channels from 0.045 ± 0.003 to 0.008 ± 0.001 (n = 8, P ≤ 0.01). Subsequent addition of arachidonic acid (10 μm), a channel opener, increased the open probability of methionine-inhibited unitary currents up to 0.43 ± 0.05 at 0 mV (n = 9, P ≤ 0.05). The results of our study provide direct evidence that the response of the human detrusor to mechanical stretch is regulated by activation of mechano-gated TREK-1 channels. Impaired mechanosensation and mechanotransduction associated with the changes in stretch-activated K2P channels may underlie myogenic bladder dysfunction in humans.
Collapse
Affiliation(s)
- Qi Lei
- Division of Urology, Department of Surgery, University of Pennsylvania, PA, USA
| | - Xiao-Qing Pan
- Division of Urology, Department of Surgery, University of Pennsylvania, PA, USA
| | | | - S Bruce Malkowicz
- Division of Urology, Department of Surgery, University of Pennsylvania, PA, USA
| | - Thomas J Guzzo
- Division of Urology, Department of Surgery, University of Pennsylvania, PA, USA
| | - Anna P Malykhina
- Division of Urology, Department of Surgery, University of Pennsylvania, PA, USA
| |
Collapse
|
36
|
Veale EL, Al-Moubarak E, Bajaria N, Omoto K, Cao L, Tucker SJ, Stevens EB, Mathie A. Influence of the N terminus on the biophysical properties and pharmacology of TREK1 potassium channels. Mol Pharmacol 2014; 85:671-81. [PMID: 24509840 DOI: 10.1124/mol.113.091199] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
TWIK-related K(+) 1 (TREK1) potassium channels are members of the two-pore domain potassium channel family and contribute to background potassium conductances in many cell types, where their activity can be regulated by a variety of physiologic and pharmacologic mediators. Fenamates such as FFA (flufenamic acid; 2-{[3-(trifluoromethyl)phenyl]amino}benzoic acid), MFA [mefenamic acid; 2-(2,3-dimethylphenyl)aminobenzoic acid], NFA [niflumic acid; 2-{[3-(trifluoromethyl)phenyl]amino}nicotinic acid], and diclofenac [2-(2-(2,6-dichlorophenylamino)phenyl)acetic acid] and the related experimental drug BL-1249 [(5,6,7,8-tetrahydro-naphthalen-1-yl)-[2-(1H-tetrazol-5-yl)-phenyl]-amine] enhance the activity of TREK1 currents, and we show that BL-1249 is the most potent of these compounds. Alternative translation initiation produces a shorter, N terminus truncated form of TREK1 with a much reduced open probability and a proposed increased permeability to sodium compared with the longer form. We show that both forms of TREK1 can be activated by fenamates and that a number of mutations that affect TREK1 channel gating occlude the action of fenamates but only in the longer form of TREK1. Furthermore, fenamates produce a marked enhancement of current through the shorter, truncated form of TREK1 and reveal a K(+)-selective channel, like the long form. These results provide insight into the mechanism of TREK1 channel activation by fenamates, and, given the role of TREK1 channels in pain, they suggest a novel analgesic mechanism for these compounds.
Collapse
Affiliation(s)
- Emma L Veale
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent, United Kingdom (E.L.V., E.A.-M., N.B., A.M.); Pfizer Neusentis, Great Abington, Cambridge, United Kingdom (K.O., L.C., E.B.S.); and Clarendon Laboratory, Department of Physics, University of Oxford, Oxford, United Kingdom (S.J.T.)
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Bagriantsev SN, Ang KH, Gallardo-Godoy A, Clark KA, Arkin MR, Renslo AR, Minor DL. A high-throughput functional screen identifies small molecule regulators of temperature- and mechano-sensitive K2P channels. ACS Chem Biol 2013; 8:1841-51. [PMID: 23738709 PMCID: PMC3747594 DOI: 10.1021/cb400289x] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
K2P (KCNK) potassium channels
generate “leak”
potassium currents that strongly influence cellular excitability and
contribute to pain, somatosensation, anesthesia, and mood. Despite
their physiological importance, K2Ps lack specific pharmacology.
Addressing this issue has been complicated by the challenges that
the leak nature of K2P currents poses for electrophysiology-based
high-throughput screening strategies. Here, we present a yeast-based
high-throughput screening assay that avoids this problem. Using a
simple growth-based functional readout, we screened a library of 106,281
small molecules and identified two new inhibitors and three new activators
of the mammalian K2P channel K2P2.1 (KCNK2, TREK-1). By combining biophysical, structure–activity,
and mechanistic analysis, we developed a dihydroacridine analogue,
ML67-33, that acts as a low micromolar, selective activator of temperature-
and mechano-sensitive K2P channels. Biophysical studies
show that ML67-33 reversibly increases channel currents by activating
the extracellular selectivity filter-based C-type gate that forms
the core gating apparatus on which a variety of diverse modulatory
inputs converge. The new K2P modulators presented here,
together with the yeast-based assay, should enable both mechanistic
and physiological studies of K2P activity and facilitate
the discovery and development of other K2P small molecule
modulators.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel L. Minor
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
94720, United States
| |
Collapse
|
38
|
Bagal SK, Brown AD, Cox PJ, Omoto K, Owen RM, Pryde DC, Sidders B, Skerratt SE, Stevens EB, Storer RI, Swain NA. Ion Channels as Therapeutic Targets: A Drug Discovery Perspective. J Med Chem 2012; 56:593-624. [DOI: 10.1021/jm3011433] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Sharan K. Bagal
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Alan D. Brown
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Peter J. Cox
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - Kiyoyuki Omoto
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Robert M. Owen
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - David C. Pryde
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Benjamin Sidders
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - Sarah E. Skerratt
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Edward B. Stevens
- Pfizer Neusentis, The
Portway Building, Granta Park, Great Abington, Cambridge, CB21
6GS, U.K
| | - R. Ian Storer
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| | - Nigel A. Swain
- Worldwide Medicinal Chemistry, Pfizer Neusentis, The Portway Building, Granta Park,
Great Abington, Cambridge, CB21 6GS, U.K
| |
Collapse
|
39
|
Petkov GV. Role of potassium ion channels in detrusor smooth muscle function and dysfunction. Nat Rev Urol 2011; 9:30-40. [PMID: 22158596 DOI: 10.1038/nrurol.2011.194] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Contraction and relaxation of the detrusor smooth muscle (DSM), which makes up the wall of the urinary bladder, facilitates the storage and voiding of urine. Several families of K(+) channels, including voltage-gated K(+) (K(V)) channels, Ca(2+)-activated K(+) (K(Ca)) channels, inward-rectifying ATP-sensitive K(+) (K(ir), K(ATP)) channels, and two-pore-domain K(+) (K(2P)) channels, are expressed and functional in DSM. They control DSM excitability and contractility by maintaining the resting membrane potential and shaping the action potentials that determine the phasic nature of contractility in this tissue. Defects in DSM K(+) channel proteins or in the molecules involved in their regulatory pathways may underlie certain forms of bladder dysfunction, such as overactive bladder. K(+) channels represent an opportunity for novel pharmacological manipulation and therapeutic intervention in human DSM. Modulation of DSM K(+) channels directly or indirectly by targeting their regulatory mechanisms has the potential to control urinary bladder function. This Review summarizes our current state of knowledge of the functional role of K(+) channels in DSM in health and disease, with special emphasis on current advancements in the field.
Collapse
Affiliation(s)
- Georgi V Petkov
- Department of Pharmaceutical and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Coker Life Sciences Building, Room 609D, 715 Sumter Street, Columbia, SC 29208, USA.
| |
Collapse
|
40
|
TRP channels in urinary bladder mechanosensation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:861-79. [PMID: 21290331 DOI: 10.1007/978-94-007-0265-3_45] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Ahn HS, dela Peña I, Kim YC, Cheong JH. 4-Chloro-7-Trifluoromethyl-10 H- Benzo[4,5]furo[3,2- b]Indole-1-Carboxylic Acid (TBIC), a Putative BK Ca Channel Opener with Uterine Relaxant Activities. Pharmacology 2011; 87:331-40. [DOI: 10.1159/000328141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 04/04/2011] [Indexed: 11/19/2022]
|
42
|
Araki I, Yoshiyama M, Kobayashi H, Mochizuki T, Du S, Okada Y, Takeda M. Emerging Families of Ion Channels Involved in Urinary Bladder Nociception. Pharmaceuticals (Basel) 2010; 3:2248-2267. [PMID: 27713353 PMCID: PMC4036652 DOI: 10.3390/ph3072248] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 06/28/2010] [Accepted: 07/15/2010] [Indexed: 01/12/2023] Open
Abstract
The expression of multiple ion channels and receptors is essential for nociceptors to detect noxious stimuli of a thermal, mechanical or chemical nature. The peripheral sensory transduction systems of the urinary bladder include sensory nerve endings, urothelial cells and others whose location is suitable for transducing mechanical and chemical stimuli. There is an increasing body of evidence implicating the Deg/ENaC and TRP channel families in the control of bladder afferent excitability under physiological and pathological conditions. Pharmacological interventions targeting these ion channels may provide a new strategy for the treatment of pathological bladder sensation and pain.
Collapse
Affiliation(s)
- Isao Araki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Mitsuharu Yoshiyama
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Hideki Kobayashi
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Tsutomu Mochizuki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| | - Shuqi Du
- Department of Urology, the 1st Affiliated Hospital, China Medical University, Shenyang, China.
| | - Yusaku Okada
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Masayuki Takeda
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
43
|
Enyedi P, Czirják G. Molecular background of leak K+ currents: two-pore domain potassium channels. Physiol Rev 2010; 90:559-605. [PMID: 20393194 DOI: 10.1152/physrev.00029.2009] [Citation(s) in RCA: 675] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Two-pore domain K(+) (K(2P)) channels give rise to leak (also called background) K(+) currents. The well-known role of background K(+) currents is to stabilize the negative resting membrane potential and counterbalance depolarization. However, it has become apparent in the past decade (during the detailed examination of the cloned and corresponding native K(2P) channel types) that this primary hyperpolarizing action is not performed passively. The K(2P) channels are regulated by a wide variety of voltage-independent factors. Basic physicochemical parameters (e.g., pH, temperature, membrane stretch) and also several intracellular signaling pathways substantially and specifically modulate the different members of the six K(2P) channel subfamilies (TWIK, TREK, TASK, TALK, THIK, and TRESK). The deep implication in diverse physiological processes, the circumscribed expression pattern of the different channels, and the interesting pharmacological profile brought the K(2P) channel family into the spotlight. In this review, we focus on the physiological roles of K(2P) channels in the most extensively investigated cell types, with special emphasis on the molecular mechanisms of channel regulation.
Collapse
Affiliation(s)
- Péter Enyedi
- Department of Physiology, Semmelweis University, Budapest, Hungary.
| | | |
Collapse
|
44
|
Tang C, Shao X, Sun B, Huang W, Qiu F, Chen Y, Shi YK, Zhang EY, Wang C, Zhao X. Anticancer mechanism of peptide P18 in human leukemia K562 cells. Org Biomol Chem 2010; 8:984-7. [DOI: 10.1039/b920762g] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
45
|
Dela Peña IC, Yoon SY, Kim SM, Lee GS, Park CS, Kim YC, Cheong JH. Inhibition of intestinal motility by the putative BK(Ca) channel opener LDD175. Arch Pharm Res 2009; 32:413-20. [PMID: 19387586 DOI: 10.1007/s12272-009-1315-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 02/17/2009] [Accepted: 02/18/2009] [Indexed: 11/30/2022]
Abstract
LDD175 (4-chloro-7-trifluoromethyl-10H-benzo[4,5]furo[3,2-b]indole-1-carboxylic acid) is a benzofuroindole compound characterized previously as a potent opener of the large conductance calcium activated (BK(Ca)) channels. Activators of the BK(Ca) channels are potential therapies for smooth muscle hyperactivity disorders. The present study investigates the influence of LDD175 on the mechanical activity of the ileum smooth muscle. LDD175 inhibited spontaneous contractions of the ileum in a concentration-dependent manner (pEC(50)=5.9 +/- 0.1) (E (max)=96 +/- 1.0% at 100 muM, n=3). It also remarkably inhibited contractions due to acetylcholine (ACh) (pEC(50)=5.3 +/- 0.1)(E (max)=97.7 +/- 2.3%, n=6) and electrical field stimulation (EFS) (pEC(50)=5.5 +/- 0.1) (E (max)=83.3 +/- 6.0%, n=6). In strips precontracted by 20 mM KCl, LDD175 significantly reduced the contractions yielding a pEC(50) of 6.1 +/- 0.1 and E (max) of 96.6 +/- 0.9%, (n=6). In 60 mM KCl, a concentration-dependent inhibition was observed with respective pEC(50) and E (max) values of 4.1 +/- 0.1 and 50.8 +/- 5.0% (n=3). BK(Ca) channel blockers iberiotoxin (IbTX) and tetraethylammonium chloride (TEA, 1 mM) attenuated the relaxative effect of LDD175 but not barium chloride (BaCl(2)), and glibenclamide (K(IR) and K(ATP) channel blockers, respectively). These data demonstrate the antispasmodic activity of LDD175 attributable to the potentiation of the BK(Ca) channels.
Collapse
|
46
|
Araki I, Du S, Kobayashi H, Sawada N, Mochizuki T, Zakoji H, Takeda M. Roles of mechanosensitive ion channels in bladder sensory transduction and overactive bladder. Int J Urol 2008; 15:681-7. [PMID: 18462357 DOI: 10.1111/j.1442-2042.2008.02052.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In the storage phase, mechanical stretch stimulates bladder afferents. These signals generate sensations and trigger voiding responses, however the precise mechanisms by which mechanical stimuli excite bladder afferents are yet to be explored. For mechanosensory transduction, the presence of mechanosensors is essential in the peripheral sensory systems including sensory nerve endings, urothelium and others. There is increasing evidence that mechanosensitive ion channels, such as degenerin/epithelial Na(+) channel (ENaC) and transient receptor potential (TRP) channel families, play key roles in the mechanosensory transduction of the urinary bladder. Pharmacological interventions targeting mechanosensitive ion channels may provide a new strategy for the treatment of bladder dysfunction.
Collapse
Affiliation(s)
- Isao Araki
- Department of Urology, University of Yamanashi Interdisciplinary Graduate School of Medicine and Engineering, Chuo, Japan.
| | | | | | | | | | | | | |
Collapse
|
47
|
Takemoto J, Masumiya H, Nunoki K, Sato T, Nakagawa H, Ikeda Y, Arai Y, Yanagisawa T. Potentiation of potassium currents by beta-adrenoceptor agonists in human urinary bladder smooth muscle cells: a possible electrical mechanism of relaxation. Pharmacology 2008; 81:251-8. [PMID: 18253064 DOI: 10.1159/000114719] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 10/10/2007] [Indexed: 11/19/2022]
Abstract
We examined the effects of beta-adrenoceptor agonists on the membrane currents of smooth muscle cells from the human urinary bladder using a whole-cell patch clamp to investigate the involvement of Ca(2+)-activated K(+) (K(Ca)) channels in relaxation by beta-adrenergic agonists. With 0.05 mmol/l EGTA in the patch pipette, depolarizing pulses evoked outward rectifying currents. Isoproterenol (1 micromol/l) significantly increased the membrane currents by 75% at +80 mV with 0.05 mmol/l EGTA pipette solution. BRL 37344 (1 micromol/l) significantly increased the membrane currents by 44% at +80 mV. Iberiotoxin (100 nmol/l) significantly decreased the membrane currents by 60% at +80 mV. In the presence of iberiotoxin, the potentiation of the outward currents by isoproterenol was greatly suppressed and, in the presence of iberiotoxin and apamin (1 micromol/l), the potentiation by isoproterenol was totally abolished. On the other hand, with 5 mmol/l EGTA pipette solution, depolarizing pulses evoked smaller outward currents. Isoproterenol (1 micromol/l) did not change the membrane currents with 5 mmol/l EGTA pipette solution. The real-time PCR analysis revealed the expression of beta(2)-adrenoceptors in the cells. These results suggest that Ca(2+)-activated and iberiotoxin- and apamin-sensitive currents via both large-conductance and small-conductance K(Ca) channels could be increased by stimulation of beta(2)-adrenoceptors.
Collapse
Affiliation(s)
- Jun Takemoto
- Department of Molecular Pharmacology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The overactive bladder is a common and distressing condition that has a significant impact on the quality of life of many people worldwide. Anticholinergics remain the first line in pharmacotherapy, however the use of these agents is hindered by adverse effects and limited efficacy. Thus there is a need for more effective treatments. Recently, there has been a move towards targeting novel pathways thought to play a role in overactivity. This review aims to provide an insight into the recent developments in pharmacotherapy of the overactive bladder. RECENT FINDINGS With recent advances in our understanding of the basic science of the overactive bladder it is becoming clear that the control of bladder functioning is far more complex than previously believed. Peripherally, a prominent role has emerged for the urothelium and the underlying suburothelium in mechanosensory control, and the role of afferent pathways in pathophysiology is increasingly recognized. SUMMARY Recent research has highlighted several potential targets for treatment of the overactive bladder, particularly within the mechanosensory pathways. With the exception of botulinum toxin, however, few new therapies have emerged showing clinical benefits. A clearer understanding of the pathophysiology of the bladder will hopefully lead to more effective and tolerated treatments.
Collapse
Affiliation(s)
- Donna J Sellers
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| | | |
Collapse
|
49
|
Affiliation(s)
- Kyu-Sung Lee
- Department of Urology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Suk Lee
- Department of Urology, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Gönczi M, Szentandrássy N, Johnson IT, Heagerty AM, Weston AH. Investigation of the role of TASK-2 channels in rat pulmonary arteries; pharmacological and functional studies following RNA interference procedures. Br J Pharmacol 2006; 147:496-505. [PMID: 16432512 PMCID: PMC1616980 DOI: 10.1038/sj.bjp.0706649] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In the present study, we investigated the ability of RNA interference technology to suppress TASK-2 potassium channel expression in human embryonic kidney (HEK293) cells stably transfected with TASK-2 cDNA and in rat isolated intact pulmonary arteries. Lipofectamine-induced transfection of a specific siRNA sequence targeted against TASK-2 resulted in a dose- and time-dependent decrease in TASK-2 channel protein expression. In siRNA-transfected cells the TASK-2 peak currents were significantly smaller than in control cells at every investigated pH, while the pH sensitivity was not altered. Using scrambled siRNA as a negative control, there were no significant changes in TASK-2 protein expression or current compared to mock-transfected cells. In TASK-2 siRNA-transfected small pulmonary arteries, but not in scrambled siRNA-treated vessels, myocyte resting membrane potential at pH 7.4 was significantly less negative and the hyperpolarisations in response to increasing pH from 6.4 to 8.4 were significantly smaller compared with control. The application of levcromakalim (10 microM), NS1619 (33 microM) and a potassium channel inhibitor cocktail (5 mM 4-aminopyridine, 10 mM tetraethylammonium chloride, 30 microM Ba2+ and 10 microM glibenclamide) had similar effects in control and in siRNA-transfected vessels. The TASK-1 (anandamide-sensitive) contribution to resting membrane potential was comparable in each group. Clofilium (100 microM) generated significantly smaller responses in transfected artery segments. These results suggest that RNA interference techniques are effective at inhibiting TASK-2 channel expression in cultured cells and in intact vessels and that TASK-2 channels have a functional role in setting the membrane potential of pulmonary artery myocytes.
Collapse
Affiliation(s)
- Mónika Gönczi
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
| | - Norbert Szentandrássy
- Department of Medicine, Stopford Building, University of Manchester, Manchester M13 9PT
| | - Ian T Johnson
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
| | - Anthony M Heagerty
- Department of Medicine, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL
| | - Arthur H Weston
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
- Author for correspondence:
| |
Collapse
|