1
|
Li J, Janin A, Patoughi M, Gaudreault N, Kis L, Moha Ou Maati H, Bossé Y, Steinberg C. Circulating Autoantibodies Targeting TREK-1 in Patients With Short-Coupled Ventricular Fibrillation. Circulation 2024; 150:1944-1954. [PMID: 39315453 DOI: 10.1161/circulationaha.124.070284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Short-coupled ventricular fibrillation (SCVF) is increasingly being recognized as a distinct primary electrical disorder and cause of otherwise unexplained cardiac arrest. However, the pathophysiology of SCVF remains largely elusive. Despite extensive genetic screening, there is no convincing evidence of a robust monogenic disease gene, thus raising the speculations for alternative pathogeneses. The role of autoimmune mechanisms in SCVF has not been investigated so far. The objective of this study was to screen for circulating autoantibodies in patients with SCVF and assess their role in arrhythmogenesis. METHODS This is a prospective, single-center, case-control study enrolling cardiac arrest survivors diagnosed with SCVF or idiopathic ventricular fibrillation (IVF) between 2019 and 2023 at the Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval Inherited Arrhythmia Clinic in Canada. Plasma samples were screened for autoantibodies targeting cardiac ion channels using peptide microarray technology. Identified target autoantibodies were then purified from pooled plasma samples for subsequent cellular electrophysiological studies. RESULTS Fourteen patients with SCVF (n=4 [29% of patients] female patients; median age, 45 years [interquartile range: 36, 59]; n=14 [100% of patients] non-Hispanic White) and 19 patients with idiopathic ventricular fibrillation (n=8 [42%] female patients; median age, 49 years [38, 57]; n=19 [100%] non-Hispanic White) were enrolled in the study and compared with 38 (n=20 [53%] female subjects; median age, 45 years [29, 66]; n=36 [95%] non-Hispanic White) sex-, age- and ethnicity-matched healthy controls. During the study period, 11 (79%) SCVF probands experienced ventricular fibrillation recurrence after a median of 4.3 months (interquartile range, 0.3-20.7). Autoantibodies targeting cardiac TREK-1 (TWIK [tandem of pore-domains in a weakly inward rectifying potassium channel]-related potassium channel 1 were identified in 7 (50%) patients with SCVF (P=0.049). Patch clamp experiments demonstrated channel-activating properties of anti-TREK-1 autoantibodies that are antagonized by quinidine in both HEK293 cells and human induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS Patients with SCVF harbor circulating autoantibodies against the cardiac TREK-1 channel. Anti-TREK-1 autoantibodies not only present the first reported biomarker for SCVF, but our functional studies also suggest a direct implication in the arrhythmogenesis of SCVF.
Collapse
Affiliation(s)
- Jin Li
- Department of Physiology, University of Bern, Switzerland (J.L.)
| | - Alexandre Janin
- Department of Molecular Medicine (A.J., M.P., N.G., Y.B.), Institut Universitaire de Cardiologie et Pneumologie de Québec
- Université Claude Bernard Lyon 1, Université de Lyon, France (A.J.)
| | - Mona Patoughi
- Department of Molecular Medicine (A.J., M.P., N.G., Y.B.), Institut Universitaire de Cardiologie et Pneumologie de Québec
| | - Nathalie Gaudreault
- Department of Molecular Medicine (A.J., M.P., N.G., Y.B.), Institut Universitaire de Cardiologie et Pneumologie de Québec
| | - Lenke Kis
- Università della Svizzera Italiana, Lugano, Switzerland (L.K.)
| | - Hamid Moha Ou Maati
- Institut des Neurosciences de Montpellier, INSERM U1298, CHU Hôpital Saint Eloi, Université de Montpellier, France (H.M.O.M.)
| | - Yohan Bossé
- Department of Molecular Medicine (A.J., M.P., N.G., Y.B.), Institut Universitaire de Cardiologie et Pneumologie de Québec
- Department of Molecular Medicine (Y.B.)
- Laval University, Quebec City, Canada (Y.B.)
| | - Christian Steinberg
- Department of Medicine (C.S.), Institut Universitaire de Cardiologie et Pneumologie de Québec
| |
Collapse
|
2
|
Bechard E, Arel E, Bride J, Louradour J, Bussy X, Elloumi A, Vigor C, Soule P, Oger C, Galano JM, Durand T, Le Guennec JY, Moha-Ou-Maati H, Demion M. Activation of hTREK-1 by polyunsaturated fatty acids involves direct interaction. Sci Rep 2024; 14:15244. [PMID: 38956407 PMCID: PMC11220079 DOI: 10.1038/s41598-024-66192-w] [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: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
TREK-1 is a mechanosensitive channel activated by polyunsaturated fatty acids (PUFAs). Its activation is supposed to be linked to changes in membrane tension following PUFAs insertion. Here, we compared the effect of 11 fatty acids and ML402 on TREK-1 channel activation using the whole cell and the inside-out configurations of the patch-clamp technique. Firstly, TREK-1 activation by PUFAs is variable and related to the variable constitutive activity of TREK-1. We observed no correlation between TREK-1 activation and acyl chain length or number of double bonds suggesting that the bilayer-couple hypothesis cannot explain by itself the activation of TREK-1 by PUFAs. The membrane fluidity measurement is not modified by PUFAs at 10 µM. The spectral shift analysis in TREK-1-enriched microsomes indicates a KD,TREK1 at 44 µM of C22:6 n-3. PUFAs display the same activation and reversible kinetics than the direct activator ML402 and activate TREK-1 in both whole-cell and inside-out configurations of patch-clamp suggesting that the binding site of PUFAs is accessible from both sides of the membrane, as for ML402. Finally, we proposed a two steps mechanism: first, insertion into the membrane, with no fluidity or curvature modifications at 10 µM, and then interaction with TREK-1 channel to open it.
Collapse
Affiliation(s)
- Emilie Bechard
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Elodie Arel
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Jamie Bride
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Julien Louradour
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Xavier Bussy
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Anis Elloumi
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Claire Vigor
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | | | - Camille Oger
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Jean-Marie Galano
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Thierry Durand
- IBMM, Université de Montpellier, UMR CNRS 5247, ENSCM, Montpellier, France
| | - Jean-Yves Le Guennec
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France
| | - Hamid Moha-Ou-Maati
- IGF, Université de Montpellier, UMR CNRS 5203, Inserm 1191, Montpellier, France
- INM, Inserm U1298, Montpellier, France
| | - Marie Demion
- PhyMedExp, Université de Montpellier, Inserm U1046, UMR CNRS 9412, CHU Arnaud de Villeneuve, Bâtiment Craste de Paulet, 370 Avenue du Doyen Gaston Giraud, 34290, Montpellier Cedex 05, France.
| |
Collapse
|
3
|
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
|
4
|
Peyronnet R, Desai A, Edelmann JC, Cameron BA, Emig R, Kohl P, Dean D. Simultaneous assessment of radial and axial myocyte mechanics by combining atomic force microscopy and carbon fibre techniques. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210326. [PMID: 36189808 PMCID: PMC9527909 DOI: 10.1098/rstb.2021.0326] [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: 03/31/2022] [Accepted: 07/21/2022] [Indexed: 11/12/2022] Open
Abstract
Cardiomyocytes sense and shape their mechanical environment, contributing to its dynamics by their passive and active mechanical properties. While axial forces generated by contracting cardiomyocytes have been amply investigated, the corresponding radial mechanics remain poorly characterized. Our aim is to simultaneously monitor passive and active forces, both axially and radially, in cardiomyocytes freshly isolated from adult mouse ventricles. To do so, we combine a carbon fibre (CF) set-up with a custom-made atomic force microscope (AFM). CF allows us to apply stretch and to record passive and active forces in the axial direction. The AFM, modified for frontal access to fit in CF, is used to characterize radial cell mechanics. We show that stretch increases the radial elastic modulus of cardiomyocytes. We further find that during contraction, cardiomyocytes generate radial forces that are reduced, but not abolished, when cells are forced to contract near isometrically. Radial forces may contribute to ventricular wall thickening during contraction, together with the dynamic re-orientation of cells and sheetlets in the myocardium. This new approach for characterizing cell mechanics allows one to obtain a more detailed picture of the balance of axial and radial mechanics in cardiomyocytes at rest, during stretch, and during contraction. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Breanne A. Cameron
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad Krozingen, University of Freiburg, 79110 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | | |
Collapse
|
5
|
Xu H, Ding Y, Qi X, Zhang ZJ, Su J. Ameliorated Neurogenesis Deficits in Dentate Gyrus May Underly the Pronounced Antidepressant Effect of TREK-1 Potassium Channel Blockade in Rats with Depressive-like Behavior. ACS Chem Neurosci 2022; 13:3068-3077. [PMID: 36269040 DOI: 10.1021/acschemneuro.2c00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Depression is considered to be the most common mental disorder and is probed by several studies that chronic mild stress contributes to depression, and fortunately, most antidepressants ameliorate depressive-like behavior accompanied with reversed hippocampal neurogenesis defects. In our present study, we confirmed that different antidepressants repaired the stress-induced neuronal and behavioral deficits by modulating adult hippocampal neurogenesis. Antidepressant treatment restored the adult hippocampal neurodegeneration, which was impaired by chronic unpredicted mild stress displaying decreased proliferation and neuronal differentiation but increased apoptosis of newly formed neurons in dentate gyrus. Notably, sucrose preference ratio significantly correlated with both neuronal differentiation proportion and newborn apoptosis proportion, suggesting a mechanistic relationship between neurogenesis and behavior. Indeed, the neotype TREK-1 potassium channel blocker expressed an earlier and pronounced antidepressant manifestation compared to the traditional selective serotonin-reuptake inhibitors fluoxetine. We therefore conclude that the administration of TREK-1 potassium channel antagonism can reverse the depressive deficits caused by chronic stress quickly via regulation of adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Hua Xu
- Department of Neurology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou, Jiangsu 213200, China.,Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yingpeng Ding
- Department of Cardiology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou 213200, Jiangsu, China
| | - Xinyang Qi
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.,The Brain Cognition and Brain Disease Institute (BCBDI), CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jianhua Su
- Department of Neurology, Affiliated Jintan Hospital of Jiangsu University, Changzhou Jintan First People's Hospital, Changzhou, Jiangsu 213200, China
| |
Collapse
|
6
|
Bae Y, Choi JH, Ryoo K, Kim A, Kwon O, Jung HG, Hwang EM, Park JY. Spadin Modulates Astrocytic Passive Conductance via Inhibition of TWIK-1/TREK-1 Heterodimeric Channels. Int J Mol Sci 2020; 21:ijms21249639. [PMID: 33348878 PMCID: PMC7765948 DOI: 10.3390/ijms21249639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Astrocytes, the most abundant cell type in the brain, are non-excitable cells and play critical roles in brain function. Mature astrocytes typically exhibit a linear current-voltage relationship termed passive conductance, which is believed to enable astrocytes to maintain potassium homeostasis in the brain. We previously demonstrated that TWIK-1/TREK-1 heterodimeric channels mainly contribute to astrocytic passive conductance. However, the molecular identity of astrocytic passive conductance is still controversial and needs to be elucidated. Here, we report that spadin, an inhibitor of TREK-1, can dramatically reduce astrocytic passive conductance in brain slices. A series of gene silencing experiments demonstrated that spadin-sensitive currents are mediated by TWIK-1/TREK-1 heterodimeric channels in cultured astrocytes and hippocampal astrocytes from brain slices. Our study clearly showed that TWIK-1/TREK-1-heterodimeric channels can act as the main molecular machinery of astrocytic passive conductance, and suggested that spadin can be used as a specific inhibitor to control astrocytic passive conductance.
Collapse
Affiliation(s)
- Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Jae Hyouk Choi
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Kanghyun Ryoo
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Ajung Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Osung Kwon
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
| | - Hyun-Gug Jung
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (J.H.C.); (A.K.)
- Correspondence: (E.M.H.); (J.-Y.P.)
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea; (Y.B.); (K.R.); (O.K.); (H.-G.J.)
- Correspondence: (E.M.H.); (J.-Y.P.)
| |
Collapse
|
7
|
Pappa AM, Liu HY, Traberg-Christensen W, Thiburce Q, Savva A, Pavia A, Salleo A, Daniel S, Owens RM. Optical and Electronic Ion Channel Monitoring from Native Human Membranes. ACS NANO 2020; 14:12538-12545. [PMID: 32469490 DOI: 10.1021/acsnano.0c01330] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Transmembrane proteins represent a major target for modulating cell activity, both in terms of therapeutics drugs and for pathogen interactions. Work on screening such therapeutics or identifying toxins has been severely limited by the lack of available methods that would give high content information on functionality (ideally multimodal) and that are suitable for high-throughput. Here, we have demonstrated a platform that is capable of multimodal (optical and electronic) screening of ligand gated ion-channel activity in human-derived membranes. The TREK-1 ion-channel was expressed within supported lipid bilayers, formed via vesicle fusion of blebs obtained from the HEK cell line overexpressing TREK-1. The resulting reconstituted native membranes were confirmed via fluorescence recovery after photobleaching to form mobile bilayers on top of films of the polymeric electroactive transducer poly(3,4-ethylenedioxythiophene) polystyrenesulfonate (PEDOT:PSS). PEDOT:PSS electrodes were then used for quantitative electrochemical impedance spectroscopy measurements of ligand-mediated TREK-1 interactions with two compounds, spadin and arachidonic acid, known to suppress and activate TREK-1 channels, respectively. PEDOT:PSS-based organic electrochemical transistors were then used for combined optical and electronic measurements of TREK-1 functionality. The technology demonstrated here is highly promising for future high-throughput screening of transmembrane protein modulators owing to the robust nature of the membrane integrated device and the highly quantitative electrical signals obtained. This is in contrast with live-cell-based electrophysiology assays (e.g., patch clamp) which compare poorly in terms of cost, usability, and compatibility with optical transduction.
Collapse
Affiliation(s)
- Anna-Maria Pappa
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Han-Yuan Liu
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Walther Traberg-Christensen
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Quentin Thiburce
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| | - Aimie Pavia
- Department of Flexible Electronics, Ecole Nationale Supérieure des Mines, CMP-EMSE, 13541 Gardanne, France
- Panaxium SAS, 13100 Aix-en-Provence, France
| | - Alberto Salleo
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, United States
| | - Susan Daniel
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, New York 14853, United States
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, CB30AS Cambridge, United Kingdom
| |
Collapse
|
8
|
Al-Shammari H, Latif N, Sarathchandra P, McCormack A, Rog-Zielinska EA, Raja S, Kohl P, Yacoub MH, Peyronnet R, Chester AH. Expression and function of mechanosensitive ion channels in human valve interstitial cells. PLoS One 2020; 15:e0240532. [PMID: 33057457 PMCID: PMC7561104 DOI: 10.1371/journal.pone.0240532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 09/28/2020] [Indexed: 11/21/2022] Open
Abstract
Background The ability of heart valve cells to respond to their mechanical environment represents a key mechanism by which the integrity and function of valve cusps is maintained. A number of different mechanotransduction pathways have been implicated in the response of valve cells to mechanical stimulation. In this study, we explore the expression pattern of several mechanosensitive ion channels (MSC) and their potential to mediate mechanosensitive responses of human valve interstitial cells (VIC). Methods MSC presence and function were probed using the patch clamp technique. Protein abundance of key MSC was evaluated by Western blotting in isolated fibroblastic VIC (VICFB) and in VIC differentiated towards myofibroblastic (VICMB) or osteoblastic (VICOB) phenotypes. Expression was compared in non-calcified and calcified human aortic valves. MSC contributions to stretch-induced collagen gene expression and to VIC migration were assessed by pharmacological inhibition of specific channels. Results Two MSC types were recorded in VICFB: potassium selective and cation non-selective channels. In keeping with functional data, the presence of both TREK-1 and Kir6.1 (potassium selective), as well as TRPM4, TRPV4 and TRPC6 (cationic non-selective) channels was confirmed in VIC at the protein level. Differentiation of VICFB into VICMB or VICOB phenotypes was associated with a lower expression of TREK-1 and Kir6.1, and a higher expression of TRPV4 and TRPC6. Differences in MSC expression were also seen in non-calcified vs calcified aortic valves where TREK-1, TRPM4 and TRPV4 expression were higher in calcified compared to control tissues. Cyclic stretch-induced expression of COL I mRNA in cultured VICFB was blocked by RN-9893, a selective inhibitor of TRPV4 channels while having no effect on the stretch-induced expression of COL III. VICFB migration was blocked with the non-specific MSC blocker streptomycin and by GSK417651A an inhibitor of TRPC6/3. Conclusion Aortic VIC express a range of MSC that play a role in functional responses of these cells to mechanical stimulation. MSC expression levels differ in calcified and non-calcified valves in ways that are in part compatible with the change in expression seen between VIC phenotypes. These changes in MSC expression, and associated alterations in the ability of VIC to respond to their mechanical environment, may form novel targets for intervention during aortic valvulopathies.
Collapse
Affiliation(s)
- Hessah Al-Shammari
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Najma Latif
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
| | | | - Ann McCormack
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
| | - Eva A. Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shahzad Raja
- Harefield Hospital, Royal Brompton & Harefield NHS Foundation Trust, Harefield, United Kingdom
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Magdi H. Yacoub
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
| | - Rémi Peyronnet
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adrian H. Chester
- National Heart & Lung Institute, Imperial College London, London, United Kingdom
- Heart Science Centre, Magdi Yacoub Institute, Harefield, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Busserolles J, Ben Soussia I, Pouchol L, Marie N, Meleine M, Devilliers M, Judon C, Schopp J, Clémenceau L, Poupon L, Chapuy E, Richard S, Noble F, Lesage F, Ducki S, Eschalier A, Lolignier S. TREK1 channel activation as a new analgesic strategy devoid of opioid adverse effects. Br J Pharmacol 2020; 177:4782-4795. [PMID: 32851651 DOI: 10.1111/bph.15243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/06/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Opioids are effective painkillers. However, their risk-benefit ratio is dampened by numerous adverse effects and opioid misuse has led to a public health crisis. Safer alternatives are required, but isolating the antinociceptive effect of opioids from their adverse effects is a pharmacological challenge because activation of the μ opioid receptor triggers both the antinociceptive and adverse effects of opioids. EXPERIMENTAL APPROACH The TREK1 potassium channel is activated downstream of μ receptor and involved in the antinociceptive activity of morphine but not in its adverse effects. Bypassing the μ opioid receptor to directly activate TREK1 could therefore be a safer analgesic strategy. KEY RESULTS We developed a selective TREK1 activator, RNE28, with antinociceptive activity in naive rodents and in models of inflammatory and neuropathic pain. This activity was lost in TREK1 knockout mice or wild-type mice treated with the TREK1 blocker spadin, showing that TREK1 is required for the antinociceptive activity of RNE28. RNE28 did not induce respiratory depression, constipation, rewarding effects, or sedation at the analgesic doses tested. CONCLUSION AND IMPLICATIONS This proof-of-concept study shows that TREK1 activators could constitute a novel class of painkillers, inspired by the mechanism of action of opioids but devoid of their adverse effects.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Ismail Ben Soussia
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Université Côte d'Azur, INSERM, Valbonne, France
| | - Laetitia Pouchol
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Nicolas Marie
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Mathieu Meleine
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Maïly Devilliers
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Céline Judon
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Julien Schopp
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Loïc Clémenceau
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Laura Poupon
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Eric Chapuy
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Serge Richard
- Centre de Recherches Biologiques, CERB, Baugy, France
| | - Florence Noble
- Neuroplasticité et thérapie des addictions, Université Paris Descartes, CNRS, Inserm, Paris, France
| | - Florian Lesage
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Université Côte d'Azur, INSERM, Valbonne, France
| | - Sylvie Ducki
- ICCF, SIGMA Clermont, Université Clermont Auvergne, CNRS, Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.,Faculté de Médecine, Institut Analgesia, Clermont-Ferrand, France
| |
Collapse
|
10
|
Darkow E, Rog-Zielinska EA, Madl J, Brandel A, Siukstaite L, Omidvar R, Kohl P, Ravens U, Römer W, Peyronnet R. The Lectin LecA Sensitizes the Human Stretch-Activated Channel TREK-1 but Not Piezo1 and Binds Selectively to Cardiac Non-myocytes. Front Physiol 2020; 11:457. [PMID: 32499717 PMCID: PMC7243936 DOI: 10.3389/fphys.2020.00457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
The healthy heart adapts continuously to a complex set of dynamically changing mechanical conditions. The mechanical environment is altered by, and contributes to, multiple cardiac diseases. Mechanical stimuli are detected and transduced by cellular mechano-sensors, including stretch-activated ion channels (SAC). The precise role of SAC in the heart is unclear, in part because there are few SAC-specific pharmacological modulators. That said, most SAC can be activated by inducers of membrane curvature. The lectin LecA is a virulence factor of Pseudomonas aeruginosa and essential for P. aeruginosa-induced membrane curvature, resulting in formation of endocytic structures and bacterial cell invasion. We investigate whether LecA modulates SAC activity. TREK-1 and Piezo1 have been selected, as they are widely expressed in the body, including cardiac tissue, and they are “canonical representatives” for the potassium selective and the cation non-selective SAC families, respectively. Live cell confocal microscopy and electron tomographic imaging were used to follow binding dynamics of LecA, and to track changes in cell morphology and membrane topology in human embryonic kidney (HEK) cells and in giant unilamellar vesicles (GUV). HEK cells were further transfected with human TREK-1 or Piezo1 constructs, and ion channel activity was recorded using the patch-clamp technique. Finally, freshly isolated cardiac cells were used for studies into cell type dependency of LecA binding. LecA (500 nM) binds within seconds to the surface of HEK cells, with highest concentration at cell-cell contact sites. Local membrane invaginations are detected in the presence of LecA, both in the plasma membrane of cells (by 17 min of LecA exposure) as well as in GUV. In HEK cells, LecA sensitizes TREK-1, but not Piezo1, to voltage and mechanical stimulation. In freshly isolated cardiac cells, LecA binds to non-myocytes, but not to ventricular or atrial cardiomyocytes. This cell type specific lack of binding is observed across cardiomyocytes from mouse, rabbit, pig, and human. Our results suggest that LecA may serve as a pharmacological tool to study SAC in a cell type-preferential manner. This could aid tissue-based research into the roles of SAC in cardiac non-myocytes.
Collapse
Affiliation(s)
- Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Eva A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Josef Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Annette Brandel
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Lina Siukstaite
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Ramin Omidvar
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Winfried Römer
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Pietri M, Djillani A, Mazella J, Borsotto M, Heurteaux C. First evidence of protective effects on stroke recovery and post-stroke depression induced by sortilin-derived peptides. Neuropharmacology 2019; 158:107715. [PMID: 31325429 DOI: 10.1016/j.neuropharm.2019.107715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 01/15/2023]
Abstract
Post-stroke depression (PSD) is the most common mood disorder following stroke with high relevance for outcome and survival of patients. The TREK-1 channel represents a crucial target in the pathogenesis of stroke and depression. Spadin and its short analog mini-spadin were reported to display potent antidepressant properties. We investigated the therapeutic effects of mini-spadin in a mouse model of focal ischemia and PSD. To activate TREK-1 and induce neuroprotection a single low dose of mini-spadin (0.03 μg/kg) was intraperitoneally injected 30 min after the onset of ischemia, once a day during 7 days post-ischemia. Then, to inhibit TREK-1 and induce antidepressant effect, the peptide was injected at higher concentration (3 μg/kg) once a day for 4 days/week until the sacrifice of animals. Electrophysiological studies showed that mini-spadin had a biphasic action on TREK-1. At low doses, the channel activity was increased whereas at higher doses it was inhibited. Mini-spadin prevented the loss of body weight and the delayed dopaminergic degeneration in substantia nigra and improved the motor and cognitive ischemia-induced deficits. Moreover, mini-spadin prevented PSD analyzed in the Forced Swim (FST) and Novelty Suppressed Feeding (NSF) tests. Finally, enhanced neurogenesis and synaptogenesis contributed to the beneficial effects of mini-spadin against stroke and PSD. This work reveals the first evidence that the modulation of TREK-1 channels in the early and chronic phases of stroke as well as the stimulation of brain plasticity by mini-spadin could play a key role in its brain protective effects against stroke and its deleterious consequences such as PSD.
Collapse
Affiliation(s)
- Mariel Pietri
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Alaeddine Djillani
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Jean Mazella
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Marc Borsotto
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
| | - Catherine Heurteaux
- Université Côte D'Azur, CNRS, IPMC, UMR7275, 660 Route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| |
Collapse
|
12
|
Shabanzadeh AP, D'Onofrio PM, Magharious M, Choi KAB, Monnier PP, Koeberle PD. Modifying PTEN recruitment promotes neuron survival, regeneration, and functional recovery after CNS injury. Cell Death Dis 2019; 10:567. [PMID: 31358730 PMCID: PMC6662832 DOI: 10.1038/s41419-019-1802-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/07/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Phosphatase and tensin homolog (PTEN) regulates apoptosis and axonal growth in the developing and adult central nervous system (CNS). Here, we show that human PTEN C-terminal PDZ interactions play a critical role in neuronal apoptosis and axon regeneration after traumatic CNS injury and stroke, highlighted by the findings that antagonizing the PDZ-motif interactions of PTEN has therapeutic applicability for these indications. Interestingly, the death-inducing function of PTEN following ischemic insult depends on a PDZ-domain interaction with MAGI-2 and MAST205, PDZ proteins that are known to recruit PTEN to the plasma membrane and stabilize its interaction with PIP3. Treatments with a human peptide that prevents PTEN association with MAGI-2 or MAST205 increased neuronal survival in multiple stroke models, in vitro. A pro-survival effect was also observed in models of retinal ischemia, optic nerve transection, and after middle cerebral artery occlusion (MCAO) in adult rats. The human PTEN peptide also improved axonal regeneration in the crushed optic nerve. Furthermore, human PTEN peptide therapy promoted functional improvement after MCAO or retinal ischemia induced via ophthalmic artery ligation. These findings show that the human peptide-based targeting of C-terminal PTEN PDZ interactions has therapeutic potential for insults of the CNS, including trauma and stroke.
Collapse
Affiliation(s)
- Alireza Pirsaraei Shabanzadeh
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, M5T 2S8, Canada
| | - Philippe Matteo D'Onofrio
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mark Magharious
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Kyung An Brian Choi
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Philippe Patrick Monnier
- Departments of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, M5T 2S8, Canada
| | - Paulo Dieter Koeberle
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Rehabilitation Science Institute, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
13
|
Canella R, Martini M, Cavicchio C, Cervellati F, Benedusi M, Valacchi G. Involvement of the TREK-1 channel in human alveolar cell membrane potential and its regulation by inhibitors of the chloride current. J Cell Physiol 2019; 234:17704-17713. [PMID: 30805940 DOI: 10.1002/jcp.28396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022]
Abstract
K+ channels of the alveolar epithelium control the driving force acting on the ionic and solvent flow through the cell membrane contributing to the maintenance of cell volume and the constitution of epithelial lining fluid. In the present work, we analyze the effect of the Cl- channel inhibitors: (4-[(2-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-inden-5-yl)oxy] butanoic acid (DCPIB) and 9-anthracenecarboxylic acid (9-AC) on the total current in a type II pneumocytes (A549 cell line) model by patch clamp, immunocytochemical, and gene knockdown techniques. We noted that DCPIB and 9-AC promote the activation of K conductance. In fact, they significantly increase the intensity of the current and shift its reversal potential to values more negative than the control. By silencing outward rectifier channel in its anoctamin 6 portion, we excluded a direct involvement of Cl- ions in modulation of IK and, by means of functional tests with its specific inhibitor spadin, we identified the TREK-1 channel as the presumable target of both drugs. As the activity of TREK-1 has a key role for the correct functioning of the alveolar epithelium, the identification of DCPIB and 9-AC molecules as its activators suggests their possible use to build new pharmacological tools for the modulation of this channel.
Collapse
Affiliation(s)
- Rita Canella
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Marta Martini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Carlotta Cavicchio
- Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| | - Franco Cervellati
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mascia Benedusi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina
| |
Collapse
|
14
|
Lamas JA, Fernández-Fernández D. Tandem pore TWIK-related potassium channels and neuroprotection. Neural Regen Res 2019; 14:1293-1308. [PMID: 30964046 PMCID: PMC6524494 DOI: 10.4103/1673-5374.253506] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
TWIK-related potassium channels (TREK) belong to a subfamily of the two-pore domain potassium channels family with three members, TREK1, TREK2 and TWIK-related arachidonic acid-activated potassium channels. The two-pore domain potassium channels is the last big family of channels being discovered, therefore it is not surprising that most of the information we know about TREK channels predominantly comes from the study of heterologously expressed channels. Notwithstanding, in this review we pay special attention to the limited amount of information available on native TREK-like channels and real neurons in relation to neuroprotection. Mainly we focus on the role of free fatty acids, lysophospholipids and other neuroprotective agents like riluzole in the modulation of TREK channels, emphasizing on how important this modulation may be for the development of new therapies against neuropathic pain, depression, schizophrenia, epilepsy, ischemia and cardiac complications.
Collapse
Affiliation(s)
- J Antonio Lamas
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| | - Diego Fernández-Fernández
- Laboratory of Neuroscience, Biomedical Research Center (CINBIO), University of Vigo, Vigo, Galicia, Spain
| |
Collapse
|
15
|
Nasr N, Faucherre A, Borsotto M, Heurteaux C, Mazella J, Jopling C, Moha Ou Maati H. Identification and characterization of two zebrafish Twik related potassium channels, Kcnk2a and Kcnk2b. Sci Rep 2018; 8:15311. [PMID: 30333618 PMCID: PMC6192994 DOI: 10.1038/s41598-018-33664-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/28/2018] [Indexed: 02/05/2023] Open
Abstract
KCNK2 is a 2 pore domain potassium channel involved in maintaining cellular membrane resting potentials. Although KCNK2 is regarded as a mechanosensitive ion channel, it can also be gated chemically. Previous research indicates that KCNK2 expression is particularly enriched in neuronal and cardiac tissues. In this respect, KCNK2 plays an important role in neuroprotection and has also been linked to cardiac arrhythmias. KCNK2 has subsequently become an attractive pharmacologic target for developing preventative/curative strategies for neuro/cardio pathophysiological conditions. Zebrafish represent an important in vivo model for rapidly analysing pharmacological compounds. We therefore sought to identify and characterise zebrafish kcnk2 to allow this model system to be incorporated into therapeutic research. Our data indicates that zebrafish possess two kcnk2 orthologs, kcnk2a and kcnk2b. Electrophysiological analysis of both zebrafish Kcnk2 orthologs shows that, like their human counterparts, they are activated by different physiological stimuli such as mechanical stretch, polyunsaturated fatty acids and intracellular acidification. Furthermore, both zebrafish Kcnk2 channels are inhibited by the human KCNK2 inhibitory peptide spadin. Taken together, our results demonstrate that both Kcnk2a and Kcnk2b share similar biophysiological and pharmacological properties to human KCNK2 and indicate that the zebrafish will be a useful model for developing KCNK2 targeting strategies.
Collapse
Affiliation(s)
- Nathalie Nasr
- IGF, CNRS, INSERM, Université de Montpellier, Labex ICST, F-34094, Montpellier, France
| | - Adèle Faucherre
- IGF, CNRS, INSERM, Université de Montpellier, Labex ICST, F-34094, Montpellier, France
| | - Marc Borsotto
- IPMC, CNRS, INSERM, Université de Nice Sophia Antipolis, Labex ICST, F-06560, Valbonne, France
| | - Catherine Heurteaux
- IPMC, CNRS, INSERM, Université de Nice Sophia Antipolis, Labex ICST, F-06560, Valbonne, France
| | - Jean Mazella
- IPMC, CNRS, INSERM, Université de Nice Sophia Antipolis, Labex ICST, F-06560, Valbonne, France
| | - Chris Jopling
- IGF, CNRS, INSERM, Université de Montpellier, Labex ICST, F-34094, Montpellier, France.
| | - Hamid Moha Ou Maati
- IGF, CNRS, INSERM, Université de Montpellier, Labex ICST, F-34094, Montpellier, France.
| |
Collapse
|
16
|
Djillani A, Pietri M, Mazella J, Heurteaux C, Borsotto M. Fighting against depression with TREK-1 blockers: Past and future. A focus on spadin. Pharmacol Ther 2018; 194:185-198. [PMID: 30291907 DOI: 10.1016/j.pharmthera.2018.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Depression is a devastating mood disorder and a leading cause of disability worldwide. Depression affects approximately one in five individuals in the world and represents heavy economic and social burdens. The neurobiological mechanisms of depression are not fully understood, but evidence highlights the role of monoamine neurotransmitter balance. Several antidepressants (ADs) are marketed to treat depression and related mood disorders. However, despite their efficacy, they remain nonspecific and unsafe because they trigger serious adverse effects. Therefore, developing new molecules for new targets in depression has become a real necessity. Eight years ago, spadin was described as a natural peptide with AD properties. This 17-amino acid peptide blocks TREK-1 channels, an original target in depression. Compared to the classical AD drugs such as fluoxetine, which requires 3-4 weeks for the AD effect to manifest, spadin acts rapidly within only 4 days of treatment. The AD properties are associated with increased neurogenesis and synaptogenesis in the brain. Despite the advantages of this fast-acting AD, the in vivo stability is weak and does not last for >7 h. The present review summarizes different strategies such as retro-inverso strategy, cyclization, and shortening the spadin sequence that has led to the development and optimization of spadin as an AD. Shortened spadin analogs present increased inhibition potency for TREK-1, an improved AD activity, and prolonged in vivo bioavailability. Finally, we also discuss about other inhibitors of TREK-1 channels with a proven efficacy in treating depression in the clinic, such as fluoxetine.
Collapse
Affiliation(s)
- Alaeddine Djillani
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France; Université Côte d'Azur, CNRS, IPMC, France
| | - Mariel Pietri
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France; Université Côte d'Azur, CNRS, IPMC, France
| | - Jean Mazella
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France; Université Côte d'Azur, CNRS, IPMC, France
| | - Catherine Heurteaux
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France; Université Côte d'Azur, CNRS, IPMC, France
| | - Marc Borsotto
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'Azur, Valbonne, France; Université Côte d'Azur, CNRS, IPMC, France.
| |
Collapse
|
17
|
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
|
18
|
Djillani A, Pietri M, Moreno S, Heurteaux C, Mazella J, Borsotto M. Shortened Spadin Analogs Display Better TREK-1 Inhibition, In Vivo Stability and Antidepressant Activity. Front Pharmacol 2017; 8:643. [PMID: 28955242 PMCID: PMC5601071 DOI: 10.3389/fphar.2017.00643] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/30/2017] [Indexed: 12/28/2022] Open
Abstract
Depression is a devastating mental disorder that affects 20% of the population worldwide. Despite their proven efficacy, antidepressants present a delayed onset of action and serious adverse effects. Seven years ago, we described spadin (PE 12-28) as a promising endogenous peptide with antidepressant activity. Spadin specifically blocks the TREK-1 channel. Previously, we showed in vivo that, spadin activity disappeared beyond 7 h after administration. In order to improve in vivo spadin stability and bioavailability, we screened spadin analogs and derivatives. From the study of spadin blood degradation products, we designed a 7 amino-acid peptide, PE 22-28. In vitro studies on hTREK-1/HEK cells by using patch-clamp technique, showed that PE 22-28 displayed a better specificity and affinity for TREK-1 channel compared to spadin, IC50 of 0.12 nM vs. 40–60 nM for spadin. In the same conditions, we also pointed out that different modifications of its N or C-terminal ends maintained or abolished TREK-1 channel activity without affecting PE 22-28 affinity. In vivo, the antidepressant properties of PE 22-28 and its derivatives were demonstrated in behavioral models of depression, such as the forced swimming test. Mice treated with spadin-analogs showed a significant reduction of the immobility time. Moreover, in the novelty suppressed feeding test after a 4-day sub-chronic treatment PE 22-28 reduced significantly the latency to eat the food pellet. PE 22-28 and its analogs were able to induce neurogenesis after only a 4-day treatment with a prominent effect of the G/A-PE 22-28. On mouse cortical neurons, PE 22-28 and its derivatives enhanced synaptogenesis measured by the increase of PSD-95 expression level. Finally, the action duration of PE 22-28 and its analogs was largely improved in comparison with that of spadin, up to 23 h instead of 7 h. Taken together, our results demonstrated that PE 22-28 and its derivatives represent other promising molecules that could be an alternative to spadin in the treatment of depression.
Collapse
Affiliation(s)
- Alaeddine Djillani
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| | - Mariel Pietri
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| | - Sébastien Moreno
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| | - Catherine Heurteaux
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| | - Jean Mazella
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| | - Marc Borsotto
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Université Côte d'AzurValbonne, France
| |
Collapse
|
19
|
Kinetic properties and adrenergic control of TREK-2-like channels in rat medial prefrontal cortex (mPFC) pyramidal neurons. Brain Res 2017; 1665:95-104. [PMID: 28438532 DOI: 10.1016/j.brainres.2017.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/03/2017] [Accepted: 04/14/2017] [Indexed: 02/01/2023]
Abstract
TREK-2-like channels were identified on the basis of electrophysiological and pharmacological tests performed on freshly isolated and enzymatically/mechanically dispersed pyramidal neurons of the rat medial prefrontal cortex (mPFC). Single-channel currents were recorded in cell-attached configuration and the impact of adrenergic receptors (α1, α2, β) stimulation on spontaneously appearing TREK-2-like channel activity was tested. The obtained results indicate that noradrenaline decreases the mean open probability of TREK-2-like channel currents by activation of β1 but not of α1- and α2-adrenergic receptors. Mean open time and channel conductance were not affected. The system of intracellular signaling pathways depends on the activation of protein kinase A. We also show that adrenergic control of TREK-2-like channel currents by adrenergic receptors was similar in pyramidal neurons isolated from young, adolescent, and adult rats. Immunofluorescent confocal scans of mPFC slices confirmed the presence of the TREK-2 protein, which was abundant in layer V pyramidal neurons. The role of TREK-2-like channel control by adrenergic receptors is discussed.
Collapse
|
20
|
Tracking Effects of SIL1 Increase: Taking a Closer Look Beyond the Consequences of Elevated Expression Level. Mol Neurobiol 2017; 55:2524-2546. [PMID: 28401474 DOI: 10.1007/s12035-017-0494-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
SIL1 acts as a co-chaperone for the major ER-resident chaperone BiP and thus plays a role in many BiP-dependent cellular functions such as protein-folding control and unfolded protein response. Whereas the increase of BiP upon cellular stress conditions is a well-known phenomenon, elevation of SIL1 under stress conditions was thus far solely studied in yeast, and different studies indicated an adverse effect of SIL1 increase. This is seemingly in contrast with the beneficial effect of SIL1 increase in surviving neurons in neurodegenerative disorders such as amyotrophic lateral sclerosis and Alzheimer's disease. Here, we addressed these controversial findings. Applying cell biological, morphological and biochemical methods, we demonstrated that SIL1 increases in various mammalian cells and neuronal tissues upon cellular stress. Investigation of heterozygous SIL1 mutant cells and tissues supported this finding. Moreover, SIL1 protein was found to be stabilized during ER stress. Increased SIL1 initiates ER stress in a concentration-dependent manner which agrees with the described adverse SIL1 effect. However, our results also suggest that protective levels are achieved by the secretion of excessive SIL1 and GRP170 and that moderately increased SIL1 also ameliorates cellular fitness under stress conditions. Our immunoprecipitation results indicate that SIL1 might act in a BiP-independent manner. Proteomic studies showed that SIL1 elevation alters the expression of proteins including crucial players in neurodegeneration, especially in Alzheimer's disease. This finding agrees with our observation of increased SIL1 immunoreactivity in surviving neurons of Alzheimer's disease autopsy cases and supports the assumption that SIL1 plays a protective role in neurodegenerative disorders.
Collapse
|
21
|
Weng W, Chen Y, Wang M, Zhuang Y, Behnisch T. Potentiation of Schaffer-Collateral CA1 Synaptic Transmission by eEF2K and p38 MAPK Mediated Mechanisms. Front Cell Neurosci 2016; 10:247. [PMID: 27826228 PMCID: PMC5078695 DOI: 10.3389/fncel.2016.00247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/10/2016] [Indexed: 12/20/2022] Open
Abstract
The elongation factor 2 kinase (eEF2K), likewise known as CaMKIII, has been demonstrated to be involved in antidepressant responses of NMDA receptor antagonists. Even so, it remains open whether direct inhibition of eEF2K without altering up-stream or other signaling pathways affects hippocampal synaptic transmission and neuronal network synchrony. Inhibition of eEF2K by the selective and potent eEF2K inhibitor A-484954 induced a fast pre-synaptically mediated enhancement of synaptic transmission and synchronization of neural network activity. The eEF2K-inhibition mediated potentiation of synaptic transmission of hippocampal CA1 neurons is most notably independent of protein synthesis and does not rely on protein kinase C, protein kinase A or mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinase 1/2. Moreover, the strengthening of synaptic transmission in the response to the inhibition of eEF2K was strongly attenuated by the inhibition of p38 MAPK. In addition, we show the involvement of barium-sensitive and more specific the TWIK-related potassium-1 (TREK-1) channels in the eEF2K-inhibition mediated potentiation of synaptic transmission. These findings reveal a novel pathway of eEF2K mediated regulation of hippocampal synaptic transmission. Further research is required to study whether such compounds could be beneficial for the development of mood disorder treatments with a fast-acting antidepressant response.
Collapse
Affiliation(s)
- Weiguang Weng
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Ying Chen
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Man Wang
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Yinghan Zhuang
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| | - Thomas Behnisch
- The Institutes of Brain Science, The State Key Laboratory of Medical Neurobiology, and the Collaborative Innovation Center for Brain Science, Fudan University Shanghai, China
| |
Collapse
|
22
|
Afzali AM, Ruck T, Herrmann AM, Iking J, Sommer C, Kleinschnitz C, Preuβe C, Stenzel W, Budde T, Wiendl H, Bittner S, Meuth SG. The potassium channels TASK2 and TREK1 regulate functional differentiation of murine skeletal muscle cells. Am J Physiol Cell Physiol 2016; 311:C583-C595. [PMID: 27488672 DOI: 10.1152/ajpcell.00363.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 08/02/2016] [Indexed: 12/18/2022]
Abstract
Two-pore domain potassium (K2P) channels influence basic cellular parameters such as resting membrane potential, cellular excitability, or intracellular Ca2+-concentration [Ca2+]i While the physiological importance of K2P channels in different organ systems (e.g., heart, central nervous system, or immune system) has become increasingly clear over the last decade, their expression profile and functional role in skeletal muscle cells (SkMC) remain largely unknown. The mouse SkMC cell line C2C12, wild-type mouse muscle tissue, and primary mouse muscle cells (PMMs) were analyzed using quantitative PCR, Western blotting, and immunohistochemical stainings as well as functional analysis including patch-clamp measurements and Ca2+ imaging. Mouse SkMC express TWIK-related acid-sensitive K+ channel (TASK) 2, TWIK-related K+ channel (TREK) 1, TREK2, and TWIK-related arachidonic acid stimulated K+ channel (TRAAK). Except TASK2 all mentioned channels were upregulated in vitro during differentiation from myoblasts to myotubes. TASK2 and TREK1 were also functionally expressed and upregulated in PMMs isolated from mouse muscle tissue. Inhibition of TASK2 and TREK1 during differentiation revealed a morphological impairment of myoblast fusion accompanied by a downregulation of maturation markers. TASK2 and TREK1 blockade led to a decreased K+ outward current and a decrease of ACh-dependent Ca2+ influx in C2C12 cells as potential underlying mechanisms. K2P-channel expression was also detected in human muscle tissue by immunohistochemistry pointing towards possible relevance for human muscle cell maturation and function. In conclusion, our findings for the first time demonstrate the functional expression of TASK2 and TREK1 in muscle cells with implications for differentiation processes warranting further investigations in physiologic and pathophysiologic scenarios.
Collapse
Affiliation(s)
- Ali M Afzali
- Department of Neurology, University of Münster, Münster, Germany
| | - Tobias Ruck
- Department of Neurology, University of Münster, Münster, Germany;
| | | | - Janette Iking
- Department of Neurology, University of Münster, Münster, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | | | - Corinna Preuβe
- Department of Neuropathology, Charité-Universitätsmedizin, Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin, Berlin, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, Münster, Germany; and
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, Münster, Germany
| |
Collapse
|
23
|
Vallée N, Lambrechts K, De Maistre S, Royal P, Mazella J, Borsotto M, Heurteaux C, Abraini J, Risso JJ, Blatteau JE. Fluoxetine Protection in Decompression Sickness in Mice is Enhanced by Blocking TREK-1 Potassium Channel with the "spadin" Antidepressant. Front Physiol 2016; 7:42. [PMID: 26909044 PMCID: PMC4755105 DOI: 10.3389/fphys.2016.00042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/29/2016] [Indexed: 01/17/2023] Open
Abstract
In mice, disseminated coagulation, inflammation, and ischemia induce neurological damage that can lead to death. These symptoms result from circulating bubbles generated by a pathogenic decompression. Acute fluoxetine treatment or the presence of the TREK-1 potassium channel increases the survival rate when mice are subjected to an experimental dive/decompression protocol. This is a paradox because fluoxetine is a blocker of TREK-1 channels. First, we studied the effects of an acute dose of fluoxetine (50 mg/kg) in wild-type (WT) and TREK-1 deficient mice (knockout homozygous KO and heterozygous HET). Then, we combined the same fluoxetine treatment with a 5-day treatment protocol with spadin, in order to specifically block TREK-1 activity (KO-like mice). KO and KO-like mice were regarded as antidepressed models. In total, 167 mice (45 WTcont 46 WTflux 30 HETflux and 46 KOflux) constituting the flux-pool and 113 supplementary mice (27 KO-like 24 WTflux2 24 KO-likeflux 21 WTcont2 17 WTno dive) constituting the spad-pool were included in this study. Only 7% of KO-TREK-1 treated with fluoxetine (KOflux) and 4% of mice treated with both spadin and fluoxetine (KO-likeflux) died from decompression sickness (DCS) symptoms. These values are much lower than those of WT control (62%) or KO-like mice (41%). After the decompression protocol, mice showed significant consumption of their circulating platelets and leukocytes. Spadin antidepressed mice were more likely to exhibit DCS. Nevertheless, mice which had both blocked TREK-1 channels and fluoxetine treatment were better protected against DCS. We conclude that the protective effect of such an acute dose of fluoxetine is enhanced when TREK-1 is inhibited. We confirmed that antidepressed models may have worse DCS outcomes, but concomitant fluoxetine treatment not only decreased DCS severity but increased the survival rate.
Collapse
Affiliation(s)
- Nicolas Vallée
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle Toulon, France
| | - Kate Lambrechts
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique OpérationnelleToulon, France; UFR STAPS, Laboratoire Motricité Humaine Education Sport Santé, Université du Sud Toulon VarLa Garde, France
| | - Sébastien De Maistre
- Hôpital d'Instruction des Armées, Service de Médecine Hyperbare et Expertise Plongée Toulon, France
| | - Perrine Royal
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle Toulon, France
| | - Jean Mazella
- Centre National de la Recherche Scientifique and Université de Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 Valbonne, France
| | - Marc Borsotto
- Centre National de la Recherche Scientifique and Université de Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 Valbonne, France
| | - Catherine Heurteaux
- Centre National de la Recherche Scientifique and Université de Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275 Valbonne, France
| | - Jacques Abraini
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique OpérationnelleToulon, France; Département d'Anesthésiologie, Université LavalQuébec, QC, Canada; Faculté de Médecine, Université de Caen NormandieCaen, France
| | - Jean-Jacques Risso
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle Toulon, France
| | - Jean-Eric Blatteau
- Institut de Recherche Biomédicale des Armées, Equipe Résidante de Recherche Subaquatique Opérationnelle Toulon, France
| |
Collapse
|
24
|
TREK1 channel blockade induces an antidepressant-like response synergizing with 5-HT1A receptor signaling. Eur Neuropsychopharmacol 2015; 25:2426-36. [PMID: 26441141 DOI: 10.1016/j.euroneuro.2015.09.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 08/07/2015] [Accepted: 09/14/2015] [Indexed: 12/28/2022]
Abstract
Current antidepressants often remain the inadequate efficacy for many depressive patients, which warrant the necessary endeavor to develop the new molecules and targets for treating depression. Recently, the two-pore domain potassium channel TREK1 has been implicated in mood regulation and TREK-1 antagonists could be the promising antidepressant. This study has screened a TREK1 blocker (SID1900) with a satisfactory blood-brain barrier permeation and bioavailability. Electrophysiological research has shown that SID1900 and the previously reported TREK1 blocker (spadin) efficiently blocked TREK-1 current in HEK293 cells and specifically blocked two-pore domain potassium channels in primary-cultured rat hippocampal neurons. SID1900 and spadin induced a significant antidepressant-like response in the rat model of chronic unpredictable mild stress (CUMS). Both two TREK1 blockers substantially increased the firing rate of 5-HT-ergic neurons in the dorsal raphe nuclei (DRN) and PFC of CUMS rats. SID1900 and spadin significantly up-regulated the expression of PKA-pCREB-BDNF signaling in DRN, hippocampus and PFC of CUMS rats, which were enhanced and reversed by a 5-HTR1A agonist (8-OH-DPAT) and antagonist (WAY100635) respectively. The present findings suggested that TREK1 channel blockers posses the substantial antidepressant-like effect and have the potential synergistic effect with 5-HT1A receptor activation through the common CREB-BDNF signal transduction.
Collapse
|
25
|
Ericson E, Wennberg Huldt C, Strömstedt M, Brodin P. A novel role of the checkpoint kinase ATR in leptin signaling. Mol Cell Endocrinol 2015; 412:257-64. [PMID: 25980679 DOI: 10.1016/j.mce.2015.04.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 03/30/2015] [Accepted: 04/27/2015] [Indexed: 11/29/2022]
Abstract
In a world with increasing incidences of obesity, it becomes critical to understand the detailed regulation of appetite. To identify novel regulators of the signaling mediated by one of the key hormones of energy homeostasis, leptin, we screened a set of compounds for their effect on the downstream Signal Transducer and Activator of Transcription 3 (STAT3) signaling. Interestingly, cells exposed to inhibitors of the Ataxia Telangiectasia and RAD3-related protein ATR increased their leptin dependent STAT3 activity. This was due to failure of the cells to induce the negative feedback mediator Suppressor of Cytokine Signaling 3 (SOCS3), suggesting that ATR has a previously unknown role in the negative feedback regulation of leptin signaling. This is an important finding not only because it sheds light on additional genes involved in leptin signaling, but also because it brings forward a new potential therapeutic intervention point for increasing leptin signaling in obese individuals.
Collapse
Affiliation(s)
- Elke Ericson
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden.
| | - Charlotte Wennberg Huldt
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Maria Strömstedt
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Peter Brodin
- Bioscience, Cardiovascular and Metabolic Diseases, AstraZeneca R&D, Pepparedsleden 1, 431 83 Mölndal, Sweden
| |
Collapse
|
26
|
Devader C, Khayachi A, Veyssière J, Moha Ou Maati H, Roulot M, Moreno S, Borsotto M, Martin S, Heurteaux C, Mazella J. In vitro and in vivo regulation of synaptogenesis by the novel antidepressant spadin. Br J Pharmacol 2015; 172:2604-17. [PMID: 25598009 DOI: 10.1111/bph.13083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/10/2014] [Accepted: 01/08/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE We have described a novel antidepressant peptide, spadin, that acts by blocking the TWIK-related-potassium channel, type 1 (TREK-1). Here, we examined possible mechanisms of action of spadin at both molecular and cellular levels. EXPERIMENTAL APPROACHES Effects of spadin were measured in primary cultures of neurons or tissues from mice injected i.v. with spadin. Western blots, qPCR, histochemical and electrophysiological techniques were used. KEY RESULTS In vitro, spadin increased neuronal membrane potential and activated both the MAPK and PI3K signalling pathways, in a time- and concentration-dependent manner. The latter pathway was involved in the protective effect of spadin against staurosporine-induced apoptosis. Also, spadin enhanced both mRNA expression and protein of two markers of synaptogenesis, the post-synaptic density protein of 95 kDalton (PSD-95) and synapsin. We confirmed these effects on synaptogenesis by the observation that spadin treatment significantly increased the proportion of mature spines in cortical neurons. Finally, in vivo injections of spadin led to a rapid increase in both mRNA expression and protein level of brain-derived neurotrophic factor (BDNF) in the hippocampus, confirming the antidepressant action of the peptide. We argue for a new role of spadin in synaptogenesis as both PSD-95 and synapsin mRNA expression and protein levels were further enhanced in the hippocampus, following treatment in vivo with the peptide. CONCLUSIONS AND IMPLICATIONS These findings provide new mechanisms of action for the rapidly acting antidepressant peptide spadin by stimulating expression of BDNF and synaptic proteins, both in vitro and in vivo.
Collapse
Affiliation(s)
- C Devader
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, UMR 7275, Université de Nice-Sophia Antipolis, Valbonne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Retroinverso analogs of spadin display increased antidepressant effects. Psychopharmacology (Berl) 2015; 232:561-74. [PMID: 25080852 PMCID: PMC4302242 DOI: 10.1007/s00213-014-3683-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 07/07/2014] [Indexed: 01/22/2023]
Abstract
RATIONALE Although depression is the most common mood disorder, only one third of patients are treated with success. Finding new targets, new drugs, and also new drug intake way are the main challenges in the depression field. Several years ago, we identified a new target with the TWIK-related potassium channel-1 (TREK-1) potassium channel, and more recently, we have discovered a peptide of 17 amino acids with antidepressant properties. This peptide, that we called spadin, can be considered as a new concept in antidepressant drug design. Spadin derives from a larger peptide resulting to a posttranslational maturation of sortilin; consequently, spadin can be considered as a natural molecule. Moreover, spadin acts more rapidly than classical antidepressants and does not induce side effects. OBJECTIVES In this work, we sought analogs of spadin displaying a better affinity on TREK-1 channels and an increased action duration. METHODS Analogs were characterized by electrophysiology measurements, by behavioral tests, and by their ability to induce neurogenesis. RESULTS We identified two retro-inverso peptides that have kept the antidepressant properties of spadin; particularly, they increased the hippocampal neurogenesis after a 4-day treatment. As spadin, these analogs did not induce side effects on either pain, epilepsy processes, or at the cardiac level. CONCLUSIONS Together, our results indicated that spadin retro-inverso peptides could represent new potent antidepressant drugs. As exemplified by spadin in the field of depression, retro-inverso strategies could represent a useful technique for developing new classes of drugs in a number of pathologies.
Collapse
|
28
|
The role of K₂p channels in anaesthesia and sleep. Pflugers Arch 2014; 467:907-16. [PMID: 25482669 PMCID: PMC4428837 DOI: 10.1007/s00424-014-1654-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/20/2022]
Abstract
Tandem two-pore potassium channels (K2Ps) have widespread expression in the central nervous system and periphery where they contribute to background membrane conductance. Some general anaesthetics promote the opening of some of these channels, enhancing potassium currents and thus producing a reduction in neuronal excitability that contributes to the transition to unconsciousness. Similarly, these channels may be recruited during the normal sleep-wake cycle as downstream effectors of wake-promoting neurotransmitters such as noradrenaline, histamine and acetylcholine. These transmitters promote K2P channel closure and thus an increase in neuronal excitability. Our understanding of the roles of these channels in sleep and anaesthesia has been largely informed by the study of mouse K2P knockout lines and what is currently predicted by in vitro electrophysiology and channel structure and gating.
Collapse
|
29
|
Moha ou Maati H, Bourcier-Lucas C, Veyssiere J, Kanzari A, Heurteaux C, Borsotto M, Haddjeri N, Lucas G. The peptidic antidepressant spadin interacts with prefrontal 5-HT4 and mGluR2 receptors in the control of serotonergic function. Brain Struct Funct 2014; 221:21-37. [DOI: 10.1007/s00429-014-0890-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 09/11/2014] [Indexed: 10/24/2022]
|
30
|
Reed A, Kohl P, Peyronnet R. Molecular candidates for cardiac stretch-activated ion channels. Glob Cardiol Sci Pract 2014; 2014:9-25. [PMID: 25405172 PMCID: PMC4220428 DOI: 10.5339/gcsp.2014.19] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 06/08/2014] [Indexed: 01/20/2023] Open
Abstract
The heart is a mechanically-active organ that dynamically senses its own mechanical environment. This environment is constantly changing, on a beat-by-beat basis, with additional modulation by respiratory activity and changes in posture or physical activity, and further overlaid with more slowly occurring physiological (e.g. pregnancy, endurance training) or pathological challenges (e.g. pressure or volume overload). Far from being a simple pump, the heart detects changes in mechanical demand and adjusts its performance accordingly, both via heart rate and stroke volume alteration. Many of the underlying regulatory processes are encoded intracardially, and are thus maintained even in heart transplant recipients. Over the last three decades, molecular substrates of cardiac mechanosensitivity have gained increasing recognition in the scientific and clinical communities. Nonetheless, the processes underlying this phenomenon are still poorly understood. Stretch-activated ion channels (SAC) have been identified as one contributor to mechanosensitive autoregulation of the heartbeat. They also appear to play important roles in the development of cardiac pathologies – most notably stretch-induced arrhythmias. As recently discovered, some established cardiac drugs act, in part at least, via mechanotransduction pathways suggesting SAC as potential therapeutic targets. Clearly, identification of the molecular substrate of cardiac SAC is of clinical importance and a number of candidate proteins have been identified. At the same time, experimental studies have revealed variable–and at times contrasting–results regarding their function. Further complication arises from the fact that many ion channels that are not classically defined as SAC, including voltage and ligand-gated ion channels, can respond to mechanical stimulation. Here, we summarise what is known about the molecular substrate of the main candidates for cardiac SAC, before identifying potential further developments in this area of translational research.
Collapse
Affiliation(s)
- Alistair Reed
- Medical Sciences Division, University of Oxford, United Kingdom
| | | | | |
Collapse
|
31
|
TREK-King the Blood–Brain-Barrier. J Neuroimmune Pharmacol 2014; 9:293-301. [DOI: 10.1007/s11481-014-9530-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/09/2014] [Indexed: 10/25/2022]
|
32
|
Hund TJ, Snyder JS, Wu X, Glynn P, Koval OM, Onal B, Leymaster ND, Unudurthi SD, Curran J, Camardo C, Wright PJ, Binkley PF, Anderson ME, Mohler PJ. β(IV)-Spectrin regulates TREK-1 membrane targeting in the heart. Cardiovasc Res 2014; 102:166-75. [PMID: 24445605 DOI: 10.1093/cvr/cvu008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS Cardiac function depends on the highly regulated and co-ordinate activity of a large ensemble of potassium channels that control myocyte repolarization. While voltage-gated K(+) channels have been well characterized in the heart, much less is known about regulation and/or targeting of two-pore K(+) channel (K(2P)) family members, despite their potential importance in modulation of heart function. METHODS AND RESULTS Here, we report a novel molecular pathway for membrane targeting of TREK-1, a mechano-sensitive K(2P) channel regulated by environmental and physical factors including membrane stretch, pH, and polyunsaturated fatty acids (e.g. arachidonic acid). We demonstrate that β(IV)-spectrin, an actin-associated protein, is co-localized with TREK-1 at the myocyte intercalated disc, associates with TREK-1 in the heart, and is required for TREK-1 membrane targeting. Mice expressing β(IV)-spectrin lacking TREK-1 binding (qv(4J)) display aberrant TREK-1 membrane localization, decreased TREK-1 activity, delayed action potential repolarization, and arrhythmia without apparent defects in localization/function of other cardiac potassium channel subunits. Finally, we report abnormal β(IV)-spectrin levels in human heart failure. CONCLUSIONS These data provide new insight into membrane targeting of TREK-1 in the heart and establish a broader role for β(IV)-spectrin in organizing functional membrane domains critical for normal heart function.
Collapse
Affiliation(s)
- Thomas J Hund
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 W. 12th Avenue, 43210 Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Heyman NS, Cowles CL, Barnett SD, Wu YY, Cullison C, Singer CA, Leblanc N, Buxton ILO. TREK-1 currents in smooth muscle cells from pregnant human myometrium. Am J Physiol Cell Physiol 2013; 305:C632-42. [PMID: 23804201 PMCID: PMC3761174 DOI: 10.1152/ajpcell.00324.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 06/24/2013] [Indexed: 01/05/2023]
Abstract
The mechanisms governing maintenance of quiescence during pregnancy remain largely unknown. The current study characterizes a stretch-activated, tetraethylammonium-insensitive K(+) current in smooth muscle cells isolated from pregnant human myometrium. This study hypothesizes that these K(+) currents can be attributed to TREK-1 and that upregulation of this channel during pregnancy assists with the maintenance of a negative cell membrane potential, conceivably contributing to uterine quiescence until full term. The results of this study demonstrate that, in pregnant human myometrial cells, outward currents at 80 mV increased from 4.8 ± 1.5 to 19.4 ± 7.5 pA/pF and from 3.0 ± 0.8 to 11.8 ± 2.7 pA/pF with application of arachidonic acid (AA) and NaHCO3, respectively, causing intracellular acidification. Similarly, outward currents were inhibited following application of 10 μM fluphenazine by 51.2 ± 9.8% after activation by AA and by 73.9 ± 4.2% after activation by NaHCO3. In human embryonic kidney (HEK-293) cells stably expressing TREK-1, outward currents at 80 mV increased from 91.0 ± 23.8 to 247.5 ± 73.3 pA/pF and from 34.8 ± 8.9 to 218.6 ± 45.0 pA/pF with application of AA and NaHCO3, respectively. Correspondingly, outward currents were inhibited 89.5 ± 2.3% by 10 μM fluphenazine following activation by AA and by 91.6 ± 3.4% following activation by NaHCO3. Moreover, currents in human myometrial cells were activated by stretch and were reduced by transfection with small interfering RNA or extracellular acidification. Understanding gestational regulation of expression and gating of TREK-1 channels could be important in determining appropriate maintenance of uterine quiescence during pregnancy.
Collapse
Affiliation(s)
- Nathanael S Heyman
- Department of Pharmacology, Center for Molecular Medicine, University of Nevada School of Medicine, Reno, Nevada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bittner S, Ruck T, Schuhmann MK, Herrmann AM, Moha ou Maati H, Bobak N, Göbel K, Langhauser F, Stegner D, Ehling P, Borsotto M, Pape HC, Nieswandt B, Kleinschnitz C, Heurteaux C, Galla HJ, Budde T, Wiendl H, Meuth SG. Endothelial TWIK-related potassium channel-1 (TREK1) regulates immune-cell trafficking into the CNS. Nat Med 2013; 19:1161-5. [PMID: 23933981 DOI: 10.1038/nm.3303] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 07/16/2013] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) is an integral part of the neurovascular unit (NVU). The NVU is comprised of endothelial cells that are interconnected by tight junctions resting on a parenchymal basement membrane ensheathed by pericytes, smooth muscle cells and a layer of astrocyte end feet. Circulating blood cells, such as leukocytes, complete the NVU. BBB disruption is common in several neurological diseases, but the molecular mechanisms involved remain largely unknown. We analyzed the role of TWIK-related potassium channel-1 (TREK1, encoded by KCNK2) in human and mouse endothelial cells and the BBB. TREK1 was downregulated in endothelial cells by treatment with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). Blocking TREK1 increased leukocyte transmigration, whereas TREK1 activation had the opposite effect. We identified altered mitogen-activated protein (MAP) kinase signaling, actin remodeling and upregulation of cellular adhesion molecules as potential mechanisms of increased migration in TREK1-deficient (Kcnk2(-/-)) cells. In Kcnk2(-/-) mice, brain endothelial cells showed an upregulation of the cellular adhesion molecules ICAM1, VCAM1 and PECAM1 and facilitated leukocyte trafficking into the CNS. Following the induction of experimental autoimmune encephalomyelitis (EAE) by immunization with a myelin oligodendrocyte protein (MOG)35-55 peptide, Kcnk2(-/-) mice showed higher EAE severity scores that were accompanied by increased cellular infiltrates in the central nervous system (CNS). The severity of EAE was attenuated in mice given the amyotrophic lateral sclerosis drug riluzole or fed a diet enriched with linseed oil (which contains the TREK-1 activating omega-3 fatty acid α-linolenic acid). These beneficial effects were reduced in Kcnk2(-/-) mice, suggesting TREK-1 activating compounds may be used therapeutically to treat diseases related to BBB dysfunction.
Collapse
Affiliation(s)
- Stefan Bittner
- 1] Department of Neurology, University of Münster, Münster, Germany. [2]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Mirkovic K, Palmersheim J, Lesage F, Wickman K. Behavioral characterization of mice lacking Trek channels. Front Behav Neurosci 2012; 6:60. [PMID: 22973213 PMCID: PMC3435516 DOI: 10.3389/fnbeh.2012.00060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/23/2012] [Indexed: 11/26/2022] Open
Abstract
Two-pore domain K+ (K2P) channels are thought to underlie background K+ conductance in many cell types. The Trek subfamily of K2P channels consists of three members, Trek1/Kcnk2, Trek2/Kcnk10, and Traak/Kcnk4, all three of which are expressed in the rodent CNS. Constitutive ablation of the Trek1 gene in mice correlates with enhanced sensitivity to ischemia and epilepsy, decreased sensitivity to the effects of inhaled anesthetics, increased sensitivity to thermal and mechanical pain, and resistance to depression. While the distribution of Trek2 mRNA in the CNS is broad, little is known about the relevance of this Trek family member to neurobiology and behavior. Here, we probed the effect of constitutive Trek2 ablation, as well as the simultaneous constitutive ablation of all three Trek family genes, in paradigms that assess motor activity, coordination, anxiety-related behavior, learning and memory, and drug-induced reward-related behavior. No differences were observed between Trek2−/− and Trek1/2/Traak−/− mice in coordination or total distance traveled in an open-field. A gender-dependent impact of Trek ablation on open-field anxiety-related behavior was observed, as female but not male Trek2−/− and Trek1/2/Traak−/− mice spent more time in, and made a greater number of entries into, the center of the open-field than wild-type counterparts. Further evaluation of anxiety-related behavior in the elevated plus maze and light/dark box, however, did not reveal a significant influence of genotype on performance for either gender. Furthermore, Trek−/− mice behaved normally in tests of learning and memory, including contextual fear conditioning and novel object recognition, and with respect to opioid-induced motor stimulation and conditioned place preference (CPP). Collectively, these data argue that despite their broad distribution in the CNS, Trek channels exert a minimal influence on a wide-range of behaviors.
Collapse
Affiliation(s)
- Kelsey Mirkovic
- Department of Pharmacology, University of Minnesota Minneapolis, MN, USA
| | | | | | | |
Collapse
|
36
|
Gil V, Gallego D, Moha Ou Maati H, Peyronnet R, Martínez-Cutillas M, Heurteaux C, Borsotto M, Jiménez M. Relative contribution of SKCa and TREK1 channels in purinergic and nitrergic neuromuscular transmission in the rat colon. Am J Physiol Gastrointest Liver Physiol 2012; 303:G412-23. [PMID: 22636169 DOI: 10.1152/ajpgi.00040.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Purinergic and nitrergic neurotransmission predominantly mediate inhibitory neuromuscular transmission in the rat colon. We studied the sensitivity of both purinergic and nitrergic pathways to spadin, a TWIK-related potassium channel 1 (TREK1) inhibitor, apamin, a small-conductance calcium-activated potassium channel blocker and 1H-[1,2,4]oxadiazolo[4,3-α]quinoxalin-1-one (ODQ), a specific inhibitor of soluble guanylate cyclase. TREK1 expression was detected by RT-PCR in the rat colon. Patch-clamp experiments were performed on cells expressing hTREK1 channels. Spadin (1 μM) reduced currents 1) in basal conditions 2) activated by stretch, and 3) with arachidonic acid (AA; 10 μM). l-Methionine (1 mM) or l-cysteine (1 mM) did not modify currents activated by AA. Microelectrode and muscle bath studies were performed on rat colon samples. l-Methionine (2 mM), apamin (1 μM), ODQ (10 μM), and N(ω)-nitro-l-arginine (l-NNA; 1 mM) depolarized smooth muscle cells and increased motility. These effects were not observed with spadin (1 μM). Purinergic and nitrergic inhibitory junction potentials (IJP) were studied by incubating the tissue with l-NNA (1 mM) or MRS2500 (1 μM). Both purinergic and nitrergic IJP were unaffected by spadin. Apamin reduced both IJP with a different potency and maximal effect for each. ODQ concentration dependently abolished nitrergic IJP without affecting purinergic IJP. Similar effects were observed in hyperpolarizations induced by sodium nitroprusside (1 μM) and nitrergic relaxations induced by electrical stimulation. We propose a pharmacological approach to characterize the pathways and function of purinergic and nitrergic neurotransmission. Nitrergic neurotransmission, which is mediated by cyclic guanosine monophosphate, is insensitive to spadin, an effective TREK1 channel inhibitor. Both purinergic and nitrergic neurotransmission are inhibited by apamin but with different relative sensitivity.
Collapse
Affiliation(s)
- V Gil
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | | | | | | | | |
Collapse
|