1
|
Rinné S, Schick F, Vowinkel K, Schütte S, Krasel C, Kauferstein S, Schäfer MKH, Kiper AK, Müller T, Decher N. Potassium channel TASK-5 forms functional heterodimers with TASK-1 and TASK-3 to break its silence. Nat Commun 2024; 15:7548. [PMID: 39215006 PMCID: PMC11364637 DOI: 10.1038/s41467-024-51288-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
TASK-5 (KCNK15) belongs to the acid-sensitive subfamily of two-pore domain potassium (K2P) channels, which includes TASK-1 and TASK-3. TASK-5 stands out as K2P channel for which there is no functional data available, since it was reported in 2001 as non-functional and thus "silent". Here we show that TASK-5 channels are indeed non-functional as homodimers, but are involved in the formation of functional channel complexes with TASK-1 and TASK-3. TASK-5 negatively modulates the surface expression of TASK channels, while the heteromeric TASK-5-containing channel complexes located at the plasma membrane are characterized by changes in single-channel conductance, Gq-coupled receptor-mediated channel inhibition, and sensitivity to TASK modulators. The unique pharmacology of TASK-1/TASK-5 heterodimers, affected by a common polymorphism in KCNK15, needs to be carefully considered in the future development of drugs targeting TASK channels. Our observations provide an access to study TASK-5 at the functional level, particularly in malignant cancers associated with KCNK15.
Collapse
Affiliation(s)
- Susanne Rinné
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Florian Schick
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Kirsty Vowinkel
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Sven Schütte
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Cornelius Krasel
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Silke Kauferstein
- Centre for Sudden Cardiac Death and Institute of Legal Medicine, University Hospital Frankfurt, Goethe-University, Frankfurt/Main, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Martin K-H Schäfer
- Institute of Anatomy and Cell Biology, Philipps University Marburg, Marburg, Germany
| | - Aytug K Kiper
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany
| | - Thomas Müller
- Bayer AG, Research & Development, Pharmaceuticals, Wuppertal, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Vegetative Physiology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Kim SS, Park J, Kim E, Hwang EM, Park JY. β-COP Suppresses the Surface Expression of the TREK2. Cells 2023; 12:1500. [PMID: 37296621 PMCID: PMC10252889 DOI: 10.3390/cells12111500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
K2P channels, also known as two-pore domain K+ channels, play a crucial role in maintaining the cell membrane potential and contributing to potassium homeostasis due to their leaky nature. The TREK, or tandem of pore domains in a weak inward rectifying K+ channel (TWIK)-related K+ channel, subfamily within the K2P family consists of mechanical channels regulated by various stimuli and binding proteins. Although TREK1 and TREK2 within the TREK subfamily share many similarities, β-COP, which was previously known to bind to TREK1, exhibits a distinct binding pattern to other members of the TREK subfamily, including TREK2 and the TRAAK (TWIK-related acid-arachidonic activated K+ channel). In contrast to TREK1, β-COP binds to the C-terminus of TREK2 and reduces its cell surface expression but does not bind to TRAAK. Furthermore, β-COP cannot bind to TREK2 mutants with deletions or point mutations in the C-terminus and does not affect the surface expression of these TREK2 mutants. These results emphasize the unique role of β-COP in regulating the surface expression of the TREK family.
Collapse
Affiliation(s)
- Seong-Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
| | - Jimin Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Republic of Korea
| | - Eunju Kim
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea;
| | - Eun Mi Hwang
- Brain Science Institute (BSI), Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea;
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Republic of Korea; (S.-S.K.); (J.P.)
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul 02841, Republic of Korea
- ASTRION, Inc., Seoul 02842, Republic of Korea
| |
Collapse
|
3
|
Nguyen NH, Brodsky JL. The cellular pathways that maintain the quality control and transport of diverse potassium channels. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194908. [PMID: 36638864 PMCID: PMC9908860 DOI: 10.1016/j.bbagrm.2023.194908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Potassium channels are multi-subunit transmembrane proteins that permit the selective passage of potassium and play fundamental roles in physiological processes, such as action potentials in the nervous system and organismal salt and water homeostasis, which is mediated by the kidney. Like all ion channels, newly translated potassium channels enter the endoplasmic reticulum (ER) and undergo the error-prone process of acquiring post-translational modifications, folding into their native conformations, assembling with other subunits, and trafficking through the secretory pathway to reach their final destinations, most commonly the plasma membrane. Disruptions in these processes can result in detrimental consequences, including various human diseases. Thus, multiple quality control checkpoints evolved to guide potassium channels through the secretory pathway and clear potentially toxic, aggregation-prone misfolded species. We will summarize current knowledge on the mechanisms underlying potassium channel quality control in the secretory pathway, highlight diseases associated with channel misfolding, and suggest potential therapeutic routes.
Collapse
Affiliation(s)
- Nga H Nguyen
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, 4249 Fifth Avenue, Pittsburgh, PA 15260, USA.
| |
Collapse
|
4
|
Kim SS, Bae Y, Kwon O, Kwon SH, Seo JB, Hwang EM, Park JY. β-COP Regulates TWIK1/TREK1 Heterodimeric Channel-Mediated Passive Conductance in Astrocytes. Cells 2022; 11:cells11203322. [PMID: 36291187 PMCID: PMC9600989 DOI: 10.3390/cells11203322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Mature astrocytes are characterized by a K+ conductance (passive conductance) that changes with a constant slope with voltage, which is involved in K+ homeostasis in the brain. Recently, we reported that the tandem of pore domains in a weak inward rectifying K+ channel (TWIK1 or KCNK1) and TWIK-related K+ channel 1 (TREK1 or KCNK2) form heterodimeric channels that mediate passive conductance in astrocytes. However, little is known about the binding proteins that regulate the function of the TWIK1/TREK1 heterodimeric channels. Here, we found that β-coat protein (COP) regulated the surface expression and activity of the TWIK1/TREK1 heterodimeric channels in astrocytes. β-COP binds directly to TREK1 but not TWIK1 in a heterologous expression system. However, β-COP also interacts with the TWIK1/TREK1 heterodimeric channel in a TREK1 dependent manner and enhances the surface expression of the heterodimeric channel in astrocytes. Consequently, it regulates TWIK1/TREK1 heterodimeric channel-mediated passive conductance in astrocytes in the mouse brain. Taken together, these results suggest that β-COP is a potential regulator of astrocytic passive conductance in the brain.
Collapse
Affiliation(s)
- Seong-Seop Kim
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Osung Kwon
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Seung-Hae Kwon
- Seoul Center, Korea Basic Science Institute (KBSI), Seoul 02841, Korea
| | - Jong Bok Seo
- Seoul Center, Korea Basic Science Institute (KBSI), Seoul 02841, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-3290-5637
| |
Collapse
|
5
|
Konakov MV, Teplov IY, Levin SG, Nenov MN. Anti-hypoxic effect of interleukin-10 in hippocampal neurons is mediated by modulation of TASK-1 and TASK-3 channels activity. Biochem Biophys Res Commun 2022; 615:17-23. [DOI: 10.1016/j.bbrc.2022.05.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/14/2022] [Indexed: 11/17/2022]
|
6
|
Lee GS, Zhang J, Wu Y, Zhou Y. 14-3-3 proteins promote synaptic localization of N-methyl d-aspartate receptors (NMDARs) in mouse hippocampal and cortical neurons. PLoS One 2021; 16:e0261791. [PMID: 34962957 PMCID: PMC8714094 DOI: 10.1371/journal.pone.0261791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/09/2021] [Indexed: 11/19/2022] Open
Abstract
One of the core pathogenic mechanisms for schizophrenia is believed to be dysfunction in glutamatergic synaptic transmissions, particularly hypofunction of N-methyl d-aspartate receptors (NMDARs). Previously we showed that 14-3-3 functional knockout mice exhibit schizophrenia-associated behaviors accompanied by reduced synaptic NMDARs in forebrain excitatory neurons. To investigate how 14-3-3 proteins regulate synaptic localization of NMDARs, here we examined changes in levels of synaptic NMDARs upon 14-3-3 inhibition in primary neurons. Expression of 14-3-3 protein inhibitor (difopein) in primary glutamatergic cortical and hippocampal neurons resulted in lower number of synaptic puncta containing NMDARs, including the GluN1, GluN2A, or GluN2B subunits. In heterologous cells, 14-3-3 proteins enhanced surface expression of these NMDAR subunits. Furthermore, we identified that 14-3-3ζ and ε isoforms interact with NMDARs via binding to GluN2A and GluN2B subunits. Taken together, our results demonstrate that 14-3-3 proteins play a critical role in NMDAR synaptic trafficking by promoting surface delivery of NMDAR subunits GluN1, GluN2A, and GluN2B. As NMDAR hypofunctionality is known to act as a convergence point for progression of symptoms of schizophrenia, further studies on these signaling pathways may help understand how dysfunction of 14-3-3 proteins can cause NMDAR hypofunctionality and lead to schizophrenia-associated behaviors.
Collapse
Affiliation(s)
- Gloria S. Lee
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Jiajing Zhang
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Yuying Wu
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| | - Yi Zhou
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, United States of America
| |
Collapse
|
7
|
Rangel-Garcia CI, Salvador C, Chavez-Garcia K, Diaz-Bello B, Lopez-Gonzalez Z, Vazquez-Cruz L, Angel Vazquez-Martinez J, Ortiz-Navarrete V, Riveros-Rosas H, Escobar LI. Identification of a unique endoplasmic retention motif in the Xenopus GIRK5 channel and its contribution to oocyte maturation. FEBS Open Bio 2021; 11:1093-1108. [PMID: 33565726 PMCID: PMC8016131 DOI: 10.1002/2211-5463.13113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/17/2020] [Accepted: 02/08/2021] [Indexed: 11/17/2022] Open
Abstract
G protein‐activated inward‐rectifying potassium (K+) channels (Kir3/GIRK) participate in cell excitability. The GIRK5 channel is present in Xenopus laevis oocytes. In an attempt to investigate the physiological role of GIRK5, we identified a noncanonical di‐arginine endoplasmic reticulum (ER) retention motif (KRXY). This retention motif is located at the N‐terminal region of GIRK5, coded by two small exons found only in X. laevis and X. tropicalis. These novel exons are expressed through use of an alternative transcription start site. Mutations in the sequence KRXY produced functional channels and induced progesterone‐independent oocyte meiotic progression. The chimeric proteins enhanced green fluorescent protein (EGFP)‐GIRK5‐WT and the EGFP‐GIRK5K13AR14A double mutant, were localized to the ER and the plasma membrane of the vegetal pole of the oocyte, respectively. Silencing of GIRK5 or blocking of this channel by external barium prevented progesterone‐induced meiotic progression. The endogenous level of GIRK5 protein decreased through oocyte stages in prophase I augmenting by progesterone. In conclusion, we have identified a unique mechanism by which the expression pattern of a K+ channel evolved to control Xenopus oocyte maturation.
Collapse
Affiliation(s)
- Claudia I Rangel-Garcia
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Carolina Salvador
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Karla Chavez-Garcia
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Beatriz Diaz-Bello
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Zinaeli Lopez-Gonzalez
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Lourdes Vazquez-Cruz
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Julio Angel Vazquez-Martinez
- Departamento de Biomedicina Molecular, Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, Mexico City, Mexico
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigacion y de Estudios Avanzados del Instituto Politecnico Nacional, Mexico City, Mexico
| | - Hector Riveros-Rosas
- Departamento de Bioquimica, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| | - Laura I Escobar
- Departamento de Fisiologia, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
8
|
Khoubza L, Chatelain FC, Feliciangeli S, Lesage F, Bichet D. Physiological roles of heteromerization: focus on the two-pore domain potassium channels. J Physiol 2021; 599:1041-1055. [PMID: 33347640 DOI: 10.1113/jp279870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
Potassium channels form the largest family of ion channels with more than 80 members involved in cell excitability and signalling. Most of them exist as homomeric channels, whereas specific conditions are required to obtain heteromeric channels. It is well established that heteromerization of voltage-gated and inward rectifier potassium channels affects their function, increasing the diversity of the native potassium currents. For potassium channels with two pore domains (K2P ), homomerization has long been considered the rule, their polymodal regulation by a wide diversity of physical and chemical stimuli being responsible for the adaptation of the leak potassium currents to cellular needs. This view has recently evolved with the accumulation of evidence of heteromerization between different K2P subunits. Several functional intragroup and intergroup heteromers have recently been identified, which contribute to the functional heterogeneity of this family. K2P heteromerization is involved in the modulation of channel expression and trafficking, promoting functional and signalling diversity. As illustrated in the Abstract Figure, heteromerization of TREK1 and TRAAK provides the cell with more possibilities of regulation. It is becoming increasingly evident that K2P heteromers contribute to important physiological functions including neuronal and cardiac excitability. Since heteromerization also affects the pharmacology of K2P channels, this understanding helps to establish K2P heteromers as new therapeutic targets for physiopathological conditions.
Collapse
Affiliation(s)
- Lamyaa Khoubza
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| | - Franck C Chatelain
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| | - Sylvain Feliciangeli
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France.,Inserm, 101 rue de Tolbiac, 75013, Paris, France
| | - Florian Lesage
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France.,Inserm, 101 rue de Tolbiac, 75013, Paris, France
| | - Delphine Bichet
- Université côte d'Azur, IPMC CNRS UMR7275, Laboratory of Excellence ICST, 660 route des Lucioles 06650 Valbonne, France
| |
Collapse
|
9
|
TASK channels: channelopathies, trafficking, and receptor-mediated inhibition. Pflugers Arch 2020; 472:911-922. [DOI: 10.1007/s00424-020-02403-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
10
|
Chen S, Feng X, Chen X, Zhuang Z, Xiao J, Fu H, Klein JD, Wang XH, Hoover RS, Eaton DC, Cai H. 14-3-3γ, a novel regulator of the large-conductance Ca 2+-activated K + channel. Am J Physiol Renal Physiol 2020; 319:F52-F62. [PMID: 32463725 DOI: 10.1152/ajprenal.00584.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
14-3-3γ is a small protein regulating its target proteins through binding to phosphorylated serine/threonine residues. Sequence analysis of large-conductance Ca2+-activated K+ (BK) channels revealed a putative 14-3-3 binding site in the COOH-terminal region. Our previous data showed that 14-3-3γ is widely expressed in the mouse kidney. Therefore, we hypothesized that 14-3-3γ has a novel role in the regulation of BK channel activity and protein expression. We used electrophysiology, Western blot analysis, and coimmunoprecipitation to examine the effects of 14-3-3γ on BK channels both in vitro and in vivo. We demonstrated the interaction of 14-3-3γ with BK α-subunits (BKα) by coimmunoprecipitation. In human embryonic kidney-293 cells stably expressing BKα, overexpression of 14-3-3γ significantly decreased BK channel activity and channel open probability. 14-3-3γ inhibited both total and cell surface BKα protein expression while enhancing ERK1/2 phosphorylation in Cos-7 cells cotransfected with flag-14-3-3γ and myc-BK. Knockdown of 14-3-3γ by siRNA transfection markedly increased BKα expression. Blockade of the ERK1/2 pathway by incubation with the MEK-specific inhibitor U0126 partially abolished 14-3-3γ-mediated inhibition of BK protein expression. Similarly, pretreatment of the lysosomal inhibitor bafilomycin A1 reversed the inhibitory effects of 14-3-3γ on BK protein expression. Furthermore, overexpression of 14-3-3γ significantly increased BK protein ubiquitination in embryonic kidney-293 cells stably expressing BKα. Additionally, 3 days of dietary K+ challenge reduced 14-3-3γ expression and ERK1/2 phosphorylation while enhancing renal BK protein expression and K+ excretion. These data suggest that 14-3-3γ modulates BK channel activity and protein expression through an ERK1/2-mediated ubiquitin-lysosomal pathway.
Collapse
Affiliation(s)
- Shan Chen
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyan Feng
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Xinxin Chen
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zhizhi Zhuang
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jia Xiao
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Haian Fu
- Department of Pharmacology, Emory University, School of Medicine, Atlanta, Georgia
| | - Janet D Klein
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Xiaonan H Wang
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Robert S Hoover
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia.,Physiology, Emory University, School of Medicine, Atlanta, Georgia
| | - Douglas C Eaton
- Physiology, Emory University, School of Medicine, Atlanta, Georgia
| | - Hui Cai
- Renal Divison, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, Georgia.,Physiology, Emory University, School of Medicine, Atlanta, Georgia
| |
Collapse
|
11
|
A lower X-gate in TASK channels traps inhibitors within the vestibule. Nature 2020; 582:443-447. [PMID: 32499642 DOI: 10.1038/s41586-020-2250-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/12/2020] [Indexed: 12/23/2022]
Abstract
TWIK-related acid-sensitive potassium (TASK) channels-members of the two pore domain potassium (K2P) channel family-are found in neurons1, cardiomyocytes2-4 and vascular smooth muscle cells5, where they are involved in the regulation of heart rate6, pulmonary artery tone5,7, sleep/wake cycles8 and responses to volatile anaesthetics8-11. K2P channels regulate the resting membrane potential, providing background K+ currents controlled by numerous physiological stimuli12-15. Unlike other K2P channels, TASK channels are able to bind inhibitors with high affinity, exceptional selectivity and very slow compound washout rates. As such, these channels are attractive drug targets, and TASK-1 inhibitors are currently in clinical trials for obstructive sleep apnoea and atrial fibrillation16. In general, potassium channels have an intramembrane vestibule with a selectivity filter situated above and a gate with four parallel helices located below; however, the K2P channels studied so far all lack a lower gate. Here we present the X-ray crystal structure of TASK-1, and show that it contains a lower gate-which we designate as an 'X-gate'-created by interaction of the two crossed C-terminal M4 transmembrane helices at the vestibule entrance. This structure is formed by six residues (243VLRFMT248) that are essential for responses to volatile anaesthetics10, neurotransmitters13 and G-protein-coupled receptors13. Mutations within the X-gate and the surrounding regions markedly affect both the channel-open probability and the activation of the channel by anaesthetics. Structures of TASK-1 bound to two high-affinity inhibitors show that both compounds bind below the selectivity filter and are trapped in the vestibule by the X-gate, which explains their exceptionally low washout rates. The presence of the X-gate in TASK channels explains many aspects of their physiological and pharmacological behaviour, which will be beneficial for the future development and optimization of TASK modulators for the treatment of heart, lung and sleep disorders.
Collapse
|
12
|
Goutierre M, Al Awabdh S, Donneger F, François E, Gomez-Dominguez D, Irinopoulou T, Menendez de la Prida L, Poncer JC. KCC2 Regulates Neuronal Excitability and Hippocampal Activity via Interaction with Task-3 Channels. Cell Rep 2019; 28:91-103.e7. [PMID: 31269453 DOI: 10.1016/j.celrep.2019.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/18/2019] [Accepted: 05/30/2019] [Indexed: 10/26/2022] Open
Abstract
KCC2 regulates neuronal transmembrane chloride gradients and thereby controls GABA signaling in the brain. KCC2 downregulation is observed in numerous neurological and psychiatric disorders. Paradoxical, excitatory GABA signaling is usually assumed to contribute to abnormal network activity underlying the pathology. We tested this hypothesis and explored the functional impact of chronic KCC2 downregulation in the rat dentate gyrus. Although the reversal potential of GABAA receptor currents is depolarized in KCC2 knockdown neurons, this shift is compensated by depolarization of the resting membrane potential. This reflects downregulation of leak potassium currents. We show KCC2 interacts with Task-3 (KCNK9) channels and is required for their membrane expression. Increased neuronal excitability upon KCC2 suppression altered dentate gyrus rhythmogenesis, which could be normalized by chemogenetic hyperpolarization. Our data reveal KCC2 downregulation engages complex synaptic and cellular alterations beyond GABA signaling that perturb network activity thus offering additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Marie Goutierre
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Sana Al Awabdh
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Florian Donneger
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Emeline François
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Daniel Gomez-Dominguez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
| | - Theano Irinopoulou
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | | | - Jean Christophe Poncer
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Université, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France.
| |
Collapse
|
13
|
Capera J, Serrano-Novillo C, Navarro-Pérez M, Cassinelli S, Felipe A. The Potassium Channel Odyssey: Mechanisms of Traffic and Membrane Arrangement. Int J Mol Sci 2019; 20:ijms20030734. [PMID: 30744118 PMCID: PMC6386995 DOI: 10.3390/ijms20030734] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/29/2022] Open
Abstract
Ion channels are transmembrane proteins that conduct specific ions across biological membranes. Ion channels are present at the onset of many cellular processes, and their malfunction triggers severe pathologies. Potassium channels (KChs) share a highly conserved signature that is necessary to conduct K⁺ through the pore region. To be functional, KChs require an exquisite regulation of their subcellular location and abundance. A wide repertoire of signatures facilitates the proper targeting of the channel, fine-tuning the balance that determines traffic and location. These signature motifs can be part of the secondary or tertiary structure of the protein and are spread throughout the entire sequence. Furthermore, the association of the pore-forming subunits with different ancillary proteins forms functional complexes. These partners can modulate traffic and activity by adding their own signatures as well as by exposing or masking the existing ones. Post-translational modifications (PTMs) add a further dimension to traffic regulation. Therefore, the fate of a KCh is not fully dependent on a gene sequence but on the balance of many other factors regulating traffic. In this review, we assemble recent evidence contributing to our understanding of the spatial expression of KChs in mammalian cells. We compile specific signatures, PTMs, and associations that govern the destination of a functional channel.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Clara Serrano-Novillo
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
14
|
Ryu J, Kim DG, Lee YS, Bae Y, Kim A, Park N, Hwang EM, Park JY. Surface expression of TTYH2 is attenuated by direct interaction with β-COP. BMB Rep 2019. [PMID: 30670146 PMCID: PMC6675250 DOI: 10.5483/bmbrep.2019.52.7.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
TTYH2 is a calcium-activated, inwardly rectifying anion channel that has been shown to be related to renal cancer and colon cancer. Based on the topological prediction, TTYH2 protein has five transmembrane domains with the extracellular N-terminus and the cytoplasmic C-terminus. In the present study, we identified a vesicle transport protein, β-COP, as a novel specific binding partner of TTYH2 by yeast two-hybrid screening using a human brain cDNA library with the C-terminal region of TTYH2 (TTYH2-C) as a bait. Using in vitro and in vivo binding assays, we confirmed the protein-protein interactions between TTYH2 and β-COP. We also found that the surface expression and activity of TTYH2 were decreased by co-expression with β-COP in the heterologous expression system. In addition, β-COP associated with TTYH2 in a native condition at a human colon cancer cell line, LoVo cells. The over-expression of β-COP in the LoVo cells led to a dramatic decrease in the surface expression and activity of endogenous TTYH2. Collectively, these data suggested that β-COP plays a critical role in the trafficking of the TTYH2 channel to the plasma membrane.
Collapse
Affiliation(s)
- Jiwon Ryu
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Dong-Gyu Kim
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea
| | - Young-Sun Lee
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Yeonju Bae
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Ajung Kim
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea; KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Nammi Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| | - Eun Mi Hwang
- Korea Institute of Science and Technology (KIST), Center for Functional Connectomics, Seoul 02792, Korea; KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
15
|
A light-gated potassium channel for sustained neuronal inhibition. Nat Methods 2018; 15:969-976. [PMID: 30377377 DOI: 10.1038/s41592-018-0186-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022]
Abstract
Currently available inhibitory optogenetic tools provide short and transient silencing of neurons, but they cannot provide long-lasting inhibition because of the requirement for high light intensities. Here we present an optimized blue-light-sensitive synthetic potassium channel, BLINK2, which showed good expression in neurons in three species. The channel is activated by illumination with low doses of blue light, and in our experiments it remained active over (tens of) minutes in the dark after the illumination was stopped. This activation caused long periods of inhibition of neuronal firing in ex vivo recordings of mouse neurons and impaired motor neuron response in zebrafish in vivo. As a proof-of-concept application, we demonstrated that in a freely moving rat model of neuropathic pain, the activation of a small number of BLINK2 channels caused a long-lasting (>30 min) reduction in pain sensation.
Collapse
|
16
|
Mackie TD, Brodsky JL. Investigating Potassium Channels in Budding Yeast: A Genetic Sandbox. Genetics 2018; 209:637-650. [PMID: 29967058 PMCID: PMC6028241 DOI: 10.1534/genetics.118.301026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/15/2018] [Indexed: 12/26/2022] Open
Abstract
Like all species, the model eukaryote Saccharomyces cerevisiae, or Bakers' yeast, concentrates potassium in the cytosol as an electrogenic osmolyte and enzyme cofactor. Yeast are capable of robust growth on a wide variety of potassium concentrations, ranging from 10 µM to 2.5 M, due to the presence of a high-affinity potassium uptake system and a battery of cation exchange transporters. Genetic perturbation of either of these systems retards yeast growth on low or high potassium, respectively. However, these potassium-sensitized yeast are a powerful genetic tool, which has been leveraged for diverse studies. Notably, the potassium-sensitive cells can be transformed with plasmids encoding potassium channels from bacteria, plants, or mammals, and subsequent changes in growth rate have been found to correlate with the activity of the introduced potassium channel. Discoveries arising from the use of this assay over the past three decades have increased our understanding of the structure-function relationships of various potassium channels, the mechanisms underlying the regulation of potassium channel function and trafficking, and the chemical basis of potassium channel modulation. In this article, we provide an overview of the major genetic tools used to study potassium channels in S. cerevisiae, a survey of seminal studies utilizing these tools, and a prospective for the future use of this elegant genetic approach.
Collapse
Affiliation(s)
- Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| |
Collapse
|
17
|
Olschewski A, Veale EL, Nagy BM, Nagaraj C, Kwapiszewska G, Antigny F, Lambert M, Humbert M, Czirják G, Enyedi P, Mathie A. TASK-1 (KCNK3) channels in the lung: from cell biology to clinical implications. Eur Respir J 2017; 50:50/5/1700754. [PMID: 29122916 DOI: 10.1183/13993003.00754-2017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/05/2017] [Indexed: 12/18/2022]
Abstract
TWIK-related acid-sensitive potassium channel 1 (TASK-1 encoded by KCNK3) belongs to the family of two-pore domain potassium channels. This gene subfamily is constitutively active at physiological resting membrane potentials in excitable cells, including smooth muscle cells, and has been particularly linked to the human pulmonary circulation. TASK-1 channels are sensitive to a wide array of physiological and pharmacological mediators that affect their activity such as unsaturated fatty acids, extracellular pH, hypoxia, anaesthetics and intracellular signalling pathways. Recent studies show that modulation of TASK-1 channels, either directly or indirectly by targeting their regulatory mechanisms, has the potential to control pulmonary arterial tone in humans. Furthermore, mutations in KCNK3 have been identified as a rare cause of both familial and idiopathic pulmonary arterial hypertension. This review summarises our current state of knowledge of the functional role of TASK-1 channels in the pulmonary circulation in health and disease, with special emphasis on current advancements in the field.
Collapse
Affiliation(s)
- Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria .,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Emma L Veale
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| | - Bence M Nagy
- Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Graz, Austria.,Institute of Physiology, Medical University of Graz, Graz, Austria
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, Kremlin-Bicêtre, France.,AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.,UMRS 999, INSERM and Univ. Paris-Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, Le Plessis Robinson, France
| | - Gábor Czirják
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Enyedi
- Dept of Physiology, Semmelweis University, Budapest, Hungary
| | - Alistair Mathie
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, UK
| |
Collapse
|
18
|
Heterodimerization of two pore domain K+ channel TASK1 and TALK2 in living heterologous expression systems. PLoS One 2017; 12:e0186252. [PMID: 29016681 PMCID: PMC5634629 DOI: 10.1371/journal.pone.0186252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/27/2017] [Indexed: 11/19/2022] Open
Abstract
Two-pore-domain K+ (K2P) channels sense a wide variety of stimuli such as mechanical stress, inhalational anesthetics, and changes in extracellular pH or temperature. The K2P channel activity forms a background K+ current and, thereby, contributes to resting membrane potentials. Six subfamilies including fifteen subtypes of K2P channels have been identified. Each K2P channel molecule with two pores consists of a homodimer of each subtype. In addition, a few heterodimers mainly within the same subfamilies have been found recently. In the present study, the possibility of heterodimerization between TASK1 (TWIK-Related Acid-Sensitive K+ channel) and TALK2 (TWIK-Related Alkaline pH-Activated K+ channel) was examined. These channels belong to separate subfamilies and show extremely different channel properties. Surprisingly, single molecular imaging analyses in this study using a total internal reflection microscope suggested the heterodimerization of TASK1 and TALK2 in a pancreatic cell line, QGP-1. This heterodimer was also detected using a bimolecular fluorescence complementation assay in a HEK293 heterologous expression system. Fluorescence resonance energy transfer analyses showed that the affinity between TASK1 and TALK2 appeared to be close to those of homodimers. Whole-cell patch-clamp recordings revealed that TASK1 currents in HEK293 cells were significantly attenuated by co-expression of a dominant-negative form of TALK2 in comparison with that of wild-type TALK2. The sensitivities of TASK1-TALK2 tandem constructs to extracellular pH and halothane were characterized as a unique hybrid of TASK1 and TALK2. These results suggested that heterodimerization of TASK1 and TALK2 provides cells with the ability to make multiple responses to a variety of physiological and pharmacological stimuli.
Collapse
|
19
|
Taguchi YH. Principal component analysis based unsupervised feature extraction applied to publicly available gene expression profiles provides new insights into the mechanisms of action of histone deacetylase inhibitors. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.nepig.2016.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Fernández-Orth J, Ehling P, Ruck T, Pankratz S, Hofmann MS, Landgraf P, Dieterich DC, Smalla KH, Kähne T, Seebohm G, Budde T, Wiendl H, Bittner S, Meuth SG. 14-3-3 Proteins regulate K 2P 5.1 surface expression on T lymphocytes. Traffic 2016; 18:29-43. [PMID: 27743426 DOI: 10.1111/tra.12455] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 01/10/2023]
Abstract
K2P 5.1 channels (also called TASK-2 or Kcnk5) have already been shown to be relevant in the pathophysiology of autoimmune disease because they are known to be upregulated on peripheral and central T lymphocytes of multiple sclerosis (MS) patients. Moreover, overexpression of K2P 5.1 channels in vitro provokes enhanced T-cell effector functions. However, the molecular mechanisms regulating intracellular K2P 5.1 channel trafficking are unknown so far. Thus, the aim of the study is to elucidate the trafficking of K2P 5.1 channels on T lymphocytes. Using mass spectrometry analysis, we have identified 14-3-3 proteins as novel binding partners of K2P 5.1 channels. We show that a non-classical 14-3-3 consensus motif (R-X-X-pT/S-x) at the channel's C-terminus allows the binding between K2P 5.1 and 14-3-3. The mutant K2P 5.1/S266A diminishes the protein-protein interaction and reduces the amplitude of membrane currents. Application of a non-peptidic 14-3-3 inhibitor (BV02) significantly reduces the number of wild-type channels in the plasma membrane, whereas the drug has no effect on the trafficking of the mutated channel. Furthermore, blocker application reduces T-cell effector functions. Taken together, we demonstrate that 14-3-3 interacts with K2P 5.1 and plays an important role in channel trafficking.
Collapse
Affiliation(s)
| | - Petra Ehling
- Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Tobias Ruck
- Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Susann Pankratz
- Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | | | - Peter Landgraf
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto von-Guericke-University, Magdeburg, Germany
| | - Daniela C Dieterich
- Neural Plasticity and Communication, Institute for Pharmacology and Toxicology, Otto von-Guericke-University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Otto von-Guericke-University, Magdeburg, Germany
| | - Karl-Heinz Smalla
- Special Lab Molecular Biological Techniques, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Thilo Kähne
- Institute of Experimental Internal Medicine, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | - Guiscard Seebohm
- Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Münster, Germany
| | - Thomas Budde
- Institute for Physiology I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Sven G Meuth
- Department of Neurology, Westfälische Wilhelms-Universität, Münster, Germany
| |
Collapse
|
21
|
δ-COP contains a helix C-terminal to its longin domain key to COPI dynamics and function. Proc Natl Acad Sci U S A 2016; 113:6916-21. [PMID: 27298352 DOI: 10.1073/pnas.1603544113] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Membrane recruitment of coatomer and formation of coat protein I (COPI)-coated vesicles is crucial to homeostasis in the early secretory pathway. The conformational dynamics of COPI during cargo capture and vesicle formation is incompletely understood. By scanning the length of δ-COP via functional complementation in yeast, we dissect the domains of the δ-COP subunit. We show that the μ-homology domain is dispensable for COPI function in the early secretory pathway, whereas the N-terminal longin domain is essential. We map a previously uncharacterized helix, C-terminal to the longin domain, that is specifically required for the retrieval of HDEL-bearing endoplasmic reticulum-luminal residents. It is positionally analogous to an unstructured linker that becomes helical and membrane-facing in the open form of the AP2 clathrin adaptor complex. Based on the amphipathic nature of the critical helix it may probe the membrane for lipid packing defects or mediate interaction with cargo and thus contribute to stabilizing membrane-associated coatomer.
Collapse
|
22
|
Topogenesis and cell surface trafficking of GPR34 are facilitated by positive-inside rule that effects through a tri-basic motif in the first intracellular loop. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1534-51. [PMID: 27086875 DOI: 10.1016/j.bbamcr.2016.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/06/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022]
Abstract
Protein folding, topogenesis and intracellular targeting of G protein-coupled receptors (GPCRs) must be precisely coordinated to ensure correct receptor localization. To elucidate how different steps of GPCR biosynthesis work together, we investigated the process of membrane topology determination and how it relates to the acquisition of cell surface trafficking competence in human GPR34. By monitoring a fused FLAG-tag and a conformation-sensitive native epitope during the expression of GPR34 mutant panel, a tri-basic motif in the first intracellular loop was identified as the key topogenic signal that dictates the orientation of transmembrane domain-1 (TM1). Charge disruption of the motif perturbed topogenic processes and resulted in the conformational epitope loss, post-translational processing alteration, and trafficking arrest in the Golgi. The placement of a cleavable N-terminal signal sequence as a surrogate topogenic determinant overcame the effects of tri-basic motif mutations and rectified the TM1 orientation; thereby restored the conformational epitope, post-translational modifications, and cell surface trafficking altogether. Progressive N-tail truncation and site-directed mutagenesis revealed that a proline-rich segment of the N-tail and all four cysteines individually located in the four separate extracellular regions must simultaneously reside in the ER lumen to muster the conformational epitope. Oxidation of all four cysteines was necessary for the epitope formation, but the cysteine residues themselves were not required for the trafficking event. The underlying biochemical properties of the conformational epitope was therefore the key to understand mechanistic processes propelled by positive-inside rule that simultaneously regulate the topogenesis and intracellular trafficking of GPR34.
Collapse
|
23
|
Kilisch M, Lytovchenko O, Arakel EC, Bertinetti D, Schwappach B. A dual phosphorylation switch controls 14-3-3-dependent cell surface expression of TASK-1. J Cell Sci 2016; 129:831-42. [PMID: 26743085 PMCID: PMC4760375 DOI: 10.1242/jcs.180182] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/29/2015] [Indexed: 11/20/2022] Open
Abstract
The transport of the K+ channels TASK-1 and TASK-3 (also known as KCNK3 and KCNK9, respectively) to the cell surface is controlled by the binding of 14-3-3 proteins to a trafficking control region at the extreme C-terminus of the channels. The current model proposes that phosphorylation-dependent binding of 14-3-3 sterically masks a COPI-binding motif. However, the direct effects of phosphorylation on COPI binding and on the binding parameters of 14-3-3 isoforms are still unknown. We find that phosphorylation of the trafficking control region prevents COPI binding even in the absence of 14-3-3, and we present a quantitative analysis of the binding of all human 14-3-3 isoforms to the trafficking control regions of TASK-1 and TASK-3. Surprisingly, the affinities of 14-3-3 proteins for TASK-1 are two orders of magnitude lower than for TASK-3. Furthermore, we find that phosphorylation of a second serine residue in the C-terminus of TASK-1 inhibits 14-3-3 binding. Thus, phosphorylation of the trafficking control region can stimulate or inhibit transport of TASK-1 to the cell surface depending on the target serine residue. Our findings indicate that control of TASK-1 trafficking by COPI, kinases, phosphatases and 14-3-3 proteins is highly dynamic. Summary: Phosphorylation of a previously neglected serine residue in the trafficking control region of TASK-1 interferes with the binding of trafficking machinery and enables complex regulation by physiological stimuli.
Collapse
Affiliation(s)
- Markus Kilisch
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, Göttingen 37073, Germany
| | - Olga Lytovchenko
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, Göttingen 37073, Germany
| | - Eric C Arakel
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, Göttingen 37073, Germany
| | | | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, Göttingen 37073, Germany Max-Planck Institute for Biophysical Chemistry, Göttingen 37077, Germany
| |
Collapse
|
24
|
Zarzycka B, Kuenemann MA, Miteva MA, Nicolaes GAF, Vriend G, Sperandio O. Stabilization of protein-protein interaction complexes through small molecules. Drug Discov Today 2015; 21:48-57. [PMID: 26434617 DOI: 10.1016/j.drudis.2015.09.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/09/2015] [Accepted: 09/25/2015] [Indexed: 12/17/2022]
Abstract
Most of the small molecules that have been identified thus far to modulate protein-protein interactions (PPIs) are inhibitors. Another promising way to interfere with PPI-associated biological processes is to promote PPI stabilization. Even though PPI stabilizers are still scarce, stabilization of PPIs by small molecules is gaining momentum and offers new pharmacological options. Therefore, we have performed a literature survey of PPI stabilization using small molecules. From this, we propose a classification of PPI stabilizers based on their binding mode and the architecture of the complex to facilitate the structure-based design of stabilizers.
Collapse
Affiliation(s)
- Barbara Zarzycka
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mélaine A Kuenemann
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France
| | - Maria A Miteva
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Gert Vriend
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Olivier Sperandio
- Université Paris Diderot, Sorbonne Paris Cité, UMRS 973 Inserm, Paris 75013, France; Inserm, U973, Paris 75013, France; Faculté de Pharmacie, CDithem, 1 rue du Prof. Laguesse, 59000 Lille, France.
| |
Collapse
|
25
|
Phosphoregulatory protein 14-3-3 facilitates SAC1 transport from the endoplasmic reticulum. Proc Natl Acad Sci U S A 2015; 112:E3199-206. [PMID: 26056309 DOI: 10.1073/pnas.1509119112] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Most secretory cargo proteins in eukaryotes are synthesized in the endoplasmic reticulum and actively exported in membrane-bound vesicles that are formed by the cytosolic coat protein complex II (COPII). COPII proteins are assisted by a variety of cargo-specific adaptor proteins required for the concentration and export of secretory proteins from the endoplasmic reticulum (ER). Adaptor proteins are key regulators of cargo export, and defects in their function may result in disease phenotypes in mammals. Here we report the role of 14-3-3 proteins as a cytosolic adaptor in mediating SAC1 transport in COPII-coated vesicles. Sac1 is a phosphatidyl inositol-4 phosphate (PI4P) lipid phosphatase that undergoes serum dependent translocation between the endoplasmic reticulum and Golgi complex and controls cellular PI4P lipid levels. We developed a cell-free COPII vesicle budding reaction to examine SAC1 exit from the ER that requires COPII and at least one additional cytosolic factor, the 14-3-3 protein. Recombinant 14-3-3 protein stimulates the packaging of SAC1 into COPII vesicles and the sorting subunit of COPII, Sec24, interacts with 14-3-3. We identified a minimal sorting motif of SAC1 that is important for 14-3-3 binding and which controls SAC1 export from the ER. This LS motif is part of a 7-aa stretch, RLSNTSP, which is similar to the consensus 14-3-3 binding sequence. Homology models, based on the SAC1 structure from yeast, predict this region to be in the exposed exterior of the protein. Our data suggest a model in which the 14-3-3 protein mediates SAC1 traffic from the ER through direct interaction with a sorting signal and COPII.
Collapse
|
26
|
Renigunta V, Schlichthörl G, Daut J. Much more than a leak: structure and function of K₂p-channels. Pflugers Arch 2015; 467:867-94. [PMID: 25791628 DOI: 10.1007/s00424-015-1703-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 11/27/2022]
Abstract
Over the last decade, we have seen an enormous increase in the number of experimental studies on two-pore-domain potassium channels (K2P-channels). The collection of reviews and original articles compiled for this special issue of Pflügers Archiv aims to give an up-to-date summary of what is known about the physiology and pathophysiology of K2P-channels. This introductory overview briefly describes the structure of K2P-channels and their function in different organs. Its main aim is to provide some background information for the 19 reviews and original articles of this special issue of Pflügers Archiv. It is not intended to be a comprehensive review; instead, this introductory overview focuses on some unresolved questions and controversial issues, such as: Do K2P-channels display voltage-dependent gating? Do K2P-channels contribute to the generation of action potentials? What is the functional role of alternative translation initiation? Do K2P-channels have one or two or more gates? We come to the conclusion that we are just beginning to understand the extremely complex regulation of these fascinating channels, which are often inadequately described as 'leak channels'.
Collapse
Affiliation(s)
- Vijay Renigunta
- Institute of Physiology and Pathophysiology, Marburg University, 35037, Marburg, Germany
| | | | | |
Collapse
|
27
|
Feliciangeli S, Chatelain FC, Bichet D, Lesage F. The family of K2P channels: salient structural and functional properties. J Physiol 2015; 593:2587-603. [PMID: 25530075 DOI: 10.1113/jphysiol.2014.287268] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/10/2014] [Indexed: 12/11/2022] Open
Abstract
Potassium channels participate in many biological functions, from ion homeostasis to generation and modulation of the electrical membrane potential. They are involved in a large variety of diseases. In the human genome, 15 genes code for K(+) channels with two pore domains (K2P ). These channels form dimers of pore-forming subunits that produce background conductances finely regulated by a range of natural and chemical effectors, including signalling lipids, temperature, pressure, pH, antidepressants and volatile anaesthetics. Since the cloning of TWIK1, the prototypical member of this family, a lot of work has been carried out on their structure and biology. These studies are still in progress, but data gathered so far show that K2P channels are central players in many processes, including ion homeostasis, hormone secretion, cell development and excitability. A growing number of studies underline their implication in physiopathological mechanisms, such as vascular and pulmonary hypertension, cardiac arrhythmias, nociception, neuroprotection and depression. This review gives a synthetic view of the most noticeable features of these channels.
Collapse
Affiliation(s)
- Sylvain Feliciangeli
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Frank C Chatelain
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Delphine Bichet
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| | - Florian Lesage
- LabEx ICST, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS and Université de Nice-Sophia Antipolis, 660 Route des Lucioles, 06560, Valbonne, France
| |
Collapse
|
28
|
The role of protein-protein interactions in the intracellular traffic of the potassium channels TASK-1 and TASK-3. Pflugers Arch 2015; 467:1105-20. [PMID: 25559843 DOI: 10.1007/s00424-014-1672-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 10/24/2022]
Abstract
The intracellular transport of membrane proteins is controlled by trafficking signals: Short peptide motifs that mediate the contact with COPI, COPII or various clathrin-associated coat proteins. In addition, many membrane proteins interact with accessory proteins that are involved in the sorting of these proteins to different intracellular compartments. In the K2P channels, TASK-1 and TASK-3, the influence of protein-protein interactions on sorting decisions has been studied in some detail. Both TASK paralogues interact with the adaptor protein 14-3-3; TASK-1 interacts, in addition, with the adaptor protein p11 (S100A10) and the endosomal SNARE protein syntaxin-8. The role of these interacting proteins in controlling the intracellular traffic of the channels and the underlying molecular mechanisms are summarised in this review. In the case of 14-3-3, the interacting protein masks a retention signal in the C-terminus of the channel; in the case of p11, the interacting protein carries a retention signal that localises the channel to the endoplasmic reticulum; and in the case of syntaxin-8, the interacting protein carries an endocytosis signal that complements an endocytosis signal of the channel. These examples illustrate some of the mechanisms by which interacting proteins may determine the itinerary of a membrane protein within a cell and suggest that the intracellular traffic of membrane proteins may be adapted to the specific functions of that protein by multiple protein-protein interactions.
Collapse
|
29
|
Kim D, Kang D. Role of K₂p channels in stimulus-secretion coupling. Pflugers Arch 2014; 467:1001-11. [PMID: 25476848 DOI: 10.1007/s00424-014-1663-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 11/30/2022]
Abstract
Two-pore domain K(+) (K2P) channels are involved in a variety of physiological processes by virtue of their high basal activity and sensitivity to various biological stimuli. One of these processes is secretion of hormones and transmitters in response to stimuli such as hypoxia, acidosis, and receptor agonists. The rise in intracellular [Ca(2+)] ([Ca(2+)]i) that is critical for the secretory event can be achieved by several mechanisms: (a) inhibition of resting (background) K(+) channels, (b) activation of Na(+)/Ca(2+)-permeable channels, and (c) release of Ca(2+) from intracellular stores. Here, we discuss the role of TASK and TREK in stimulus-secretion mechanisms in carotid body chemoreceptor cells and adrenal medullary/cortical cells. Studies show that stimuli such as hypoxia and acidosis cause cell depolarization and transmitter/hormone secretion by inhibition of TASK or TREK. Subsequent elevation of [Ca(2+)]i produced by opening of voltage-dependent Ca(2+) channels then activates a Na(+)-permeable cation channel, presumably to help sustain the depolarization and [Ca(2+)]i. Agonists such as angiotensin II may elevate [Ca(2+)]i via multiple mechanisms involving both inhibition of TASK/TREK and Ca(2+) release from internal stores to cause aldosterone secretion. Thus, inhibition of resting (background) K(+) channels and subsequent activation of voltage-gated Ca(2+) channels and Na(+)-permeable non-selective cation channels may be a common ionic mechanism that lead to hormone and transmitter secretion.
Collapse
Affiliation(s)
- Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA,
| | | |
Collapse
|
30
|
O'Kelly I. Endocytosis as a mode to regulate functional expression of two-pore domain potassium (K₂p) channels. Pflugers Arch 2014; 467:1133-42. [PMID: 25413469 PMCID: PMC4428836 DOI: 10.1007/s00424-014-1641-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 10/22/2014] [Accepted: 10/24/2014] [Indexed: 11/06/2022]
Abstract
Two-pore domain potassium (K2P) channels are implicated in an array of physiological and pathophysiological roles. As a result of their biophysical properties, these channels produce a background leak K+ current which has a direct effect on cellular membrane potential and activity. The regulation of potassium leak from cells through K2P channels is of critical importance to cell function, development and survival. Controlling the cell surface expression of these channels is one mode to regulate their function and is achieved through a balance between regulated channel delivery to and retrieval from the cell surface. Here, we explore the modes of retrieval of K2P channels from the plasma membrane and observe that K2P channels are endocytosed in both a clathrin-mediated and clathrin-independent manner. K2P channels use a variety of pathways and show altered internalisation and sorting in response to external cues. These pathways working in concert, equip the cell with a range of approaches to maintain steady state levels of channels and to respond rapidly should changes in channel density be required.
Collapse
Affiliation(s)
- Ita O'Kelly
- Human Development and Health, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK, I.M.O'
| |
Collapse
|
31
|
K₂p channels in plants and animals. Pflugers Arch 2014; 467:1091-104. [PMID: 25369776 DOI: 10.1007/s00424-014-1638-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/18/2014] [Accepted: 10/21/2014] [Indexed: 10/24/2022]
Abstract
Two-pore domain potassium (K2P) channels are membrane proteins widely identified in mammals, plants, and other organisms. A functional channel is a dimer with each subunit comprising two pore-forming loops and four transmembrane domains. The genome of the model plant Arabidopsis thaliana harbors five genes coding for K2P channels. Homologs of Arabidopsis K2P channels have been found in all higher plants sequenced so far. As with the K2P channels in mammals, plant K2P channels are targets of external and internal stimuli, which fine-tune the electrical properties of the membrane for specialized transport and/or signaling tasks. Plant K2P channels are modulated by signaling molecules such as intracellular H(+) and calcium and physical factors like temperature and pressure. In this review, we ask the following: What are the similarities and differences between K2P channels in plants and animals in terms of their physiology? What is the nature of the last common ancestor (LCA) of these two groups of proteins? To answer these questions, we present physiological, structural, and phylogenetic evidence that discards the hypothesis proposing that the duplication and fusion that gave rise to the K2P channels occurred in a prokaryote LCA. Conversely, we argue that the K2P LCA was most likely a eukaryote organism. Consideration of plant and animal K2P channels in the same study is novel and likely to stimulate further exchange of ideas between students of these fields.
Collapse
|
32
|
Silent but not dumb: how cellular trafficking and pore gating modulate expression of TWIK1 and THIK2. Pflugers Arch 2014; 467:1121-31. [PMID: 25339226 DOI: 10.1007/s00424-014-1631-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/09/2014] [Accepted: 10/12/2014] [Indexed: 12/25/2022]
Abstract
Among K2P channels, a few of them turned out to be difficult to express in heterologous systems and were coined "silent subunits". Recent studies have shed light on the mechanisms behind this apparent lack of channel activity at the plasma membrane. For TWIK1 and THIK2 channels, silence is related to a combination of intracellular retention and low intrinsic activity. TWIK1 is constitutively endocytosed from the plasma membrane before being transported to recycling endosomes, whereas THIK2 is restricted to endoplasmic reticulum. These intracellular localizations are related to trafficking signals located in the cytoplasmic parts of the channels. When these motifs are mutated or masked, channels are redistributed at the plasma membrane and produce measurable currents. However, these currents are of modest amplitude. This weak basal activity is due to a hydrophobic barrier in the deep pore that limits water and ions in the conduction pathway. Other silent channels KCNK7, TWIK2, and TASK5 are still under study. Expression and characterization of these K2P channels pave the way for a better understanding of the mechanisms controlling intracellular trafficking of membrane proteins, ion conduction, and channel gating.
Collapse
|
33
|
Affiliation(s)
- Patrick E MacDonald
- University of Alberta, Department of Pharmacology and Alberta Diabetes Institute, Edmonton, Alberta, Canada T6G 2E1
| |
Collapse
|
34
|
Smyth JW, Zhang SS, Sanchez JM, Lamouille S, Vogan JM, Hesketh GG, Hong T, Tomaselli GF, Shaw RM. A 14-3-3 mode-1 binding motif initiates gap junction internalization during acute cardiac ischemia. Traffic 2014; 15:684-99. [PMID: 24612377 PMCID: PMC4278178 DOI: 10.1111/tra.12169] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 03/05/2014] [Accepted: 03/05/2014] [Indexed: 02/03/2023]
Abstract
Altered phosphorylation and trafficking of connexin 43 (Cx43) during acute ischemia contributes to arrhythmogenic gap junction remodeling, yet the critical sequence and accessory proteins necessary for Cx43 internalization remain unresolved. 14-3-3 proteins can regulate protein trafficking, and a 14-3-3 mode-1 binding motif is activated upon phosphorylation of Ser373 of the Cx43 C-terminus. We hypothesized that Cx43(Ser373) phosphorylation is important to pathological gap junction remodeling. Immunofluorescence in human heart reveals the enrichment of 14-3-3 proteins at intercalated discs, suggesting interaction with gap junctions. Knockdown of 14-3-3τ in cell lines increases gap junction plaque size at cell-cell borders. Cx43(S373A) mutation prevents Cx43/14-3-3 complexing and stabilizes Cx43 at the cell surface, indicating avoidance of degradation. Using Langendorff-perfused mouse hearts, we detect phosphorylation of newly internalized Cx43 at Ser373 and Ser368 within 30 min of no-flow ischemia. Phosphorylation of Cx43 at Ser368 by protein kinase C and Ser255 by mitogen-activated protein kinase has previously been implicated in Cx43 internalization. The Cx43(S373A) mutant is resistant to phosphorylation at both these residues and does not undergo ubiquitination, revealing Ser373 phosphorylation as an upstream gatekeeper of a posttranslational modification cascade necessary for Cx43 internalization. Cx43(Ser373) phosphorylation is a potent target for therapeutic interventions to preserve gap junction coupling in the stressed myocardium.
Collapse
Affiliation(s)
- James W. Smyth
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center
| | - Shan-Shan Zhang
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center
| | - Jose M. Sanchez
- Department of Medicine, University of California San Francisco
| | - Samy Lamouille
- Department of Medicine, University of California San Francisco
| | - Jacob M. Vogan
- Department of Medicine, University of California San Francisco
| | | | - TingTing Hong
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center
- Department of Medicine, University of California Los Angeles
| | | | - Robin M. Shaw
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center
- Department of Medicine, University of California Los Angeles
| |
Collapse
|
35
|
Enyedi P, Veres I, Braun G, Czirják G. Tubulin binds to the cytoplasmic loop of TRESK background K⁺ channel in vitro. PLoS One 2014; 9:e97854. [PMID: 24830385 PMCID: PMC4022642 DOI: 10.1371/journal.pone.0097854] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/25/2014] [Indexed: 12/02/2022] Open
Abstract
The cytoplasmic loop between the second and third transmembrane segments is pivotal in the regulation of TRESK (TWIK-related spinal cord K+ channel, K2P18.1, KCNK18). Calcineurin binds to this region and activates the channel by dephosphorylation in response to the calcium signal. Phosphorylation-dependent anchorage of 14-3-3 adaptor protein also modulates TRESK at this location. In the present study, we identified molecular interacting partners of the intracellular loop. By an affinity chromatography approach using the cytoplasmic loop as bait, we have verified the specific association of calcineurin and 14-3-3 to the channel. In addition to these known interacting proteins, we observed substantial binding of tubulin to the intracellular loop. Successive truncation of the polypeptide and pull-down experiments from mouse brain cytosol narrowed down the region sufficient for the binding of tubulin to a 16 amino acid sequence: LVLGRLSYSIISNLDE. The first six residues of this sequence are similar to the previously reported tubulin-binding region of P2X2 purinergic receptor. The tubulin-binding site of TRESK is located close to the protein kinase A (PKA)-dependent 14-3-3-docking motif of the channel. We provide experimental evidence suggesting that 14-3-3 competes with tubulin for the binding to the cytoplasmic loop of TRESK. It is intriguing that the 16 amino acid tubulin-binding sequence includes the serines, which were previously shown to be phosphorylated by microtubule-affinity regulating kinases (MARK kinases) and contribute to channel inhibition. Although tubulin binds to TRESK in vitro, it remains to be established whether the two proteins also interact in the living cell.
Collapse
Affiliation(s)
- Péter Enyedi
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Irén Veres
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gabriella Braun
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gábor Czirják
- Department of Physiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
36
|
Renigunta V, Fischer T, Zuzarte M, Kling S, Zou X, Siebert K, Limberg MM, Rinné S, Decher N, Schlichthörl G, Daut J. Cooperative endocytosis of the endosomal SNARE protein syntaxin-8 and the potassium channel TASK-1. Mol Biol Cell 2014; 25:1877-91. [PMID: 24743596 PMCID: PMC4055267 DOI: 10.1091/mbc.e13-10-0592] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
SNARE proteins can have functions unrelated to membrane fusion. The unassembled form of the SNARE protein syntaxin-8 interacts with the K+ channel TASK-1; both proteins are internalized via clathrin-mediated endocytosis in a cooperative manner. This is a novel mechanism for the control of endocytosis by cargo proteins. The endosomal SNARE protein syntaxin-8 interacts with the acid-sensitive potassium channel TASK-1. The functional relevance of this interaction was studied by heterologous expression of these proteins (and mutants thereof) in Xenopus oocytes and in mammalian cell lines. Coexpression of syntaxin-8 caused a fourfold reduction in TASK-1 current, a corresponding reduction in the expression of TASK-1 at the cell surface, and a marked increase in the rate of endocytosis of the channel. TASK-1 and syntaxin-8 colocalized in the early endosomal compartment, as indicated by the endosomal markers 2xFYVE and rab5. The stimulatory effect of the SNARE protein on the endocytosis of the channel was abolished when both an endocytosis signal in TASK-1 and an endocytosis signal in syntaxin-8 were mutated. A syntaxin-8 mutant that cannot assemble with other SNARE proteins had virtually the same effect as wild-type syntaxin-8. Total internal reflection fluorescence microscopy showed formation and endocytosis of vesicles containing fluorescence-tagged clathrin, TASK-1, and/or syntaxin-8. Our results suggest that the unassembled form of syntaxin-8 and the potassium channel TASK-1 are internalized via clathrin-mediated endocytosis in a cooperative manner. This implies that syntaxin-8 regulates the endocytosis of TASK-1. Our study supports the idea that endosomal SNARE proteins can have functions unrelated to membrane fusion.
Collapse
Affiliation(s)
- Vijay Renigunta
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Thomas Fischer
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Marylou Zuzarte
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Stefan Kling
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Xinle Zou
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Kai Siebert
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Maren M Limberg
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Niels Decher
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Günter Schlichthörl
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| | - Jürgen Daut
- Institute of Physiology and Pathophysiology, Marburg University, 35037 Marburg, Germany
| |
Collapse
|
37
|
Arakel EC, Brandenburg S, Uchida K, Zhang H, Lin YW, Kohl T, Schrul B, Sulkin MS, Efimov IR, Nichols CG, Lehnart SE, Schwappach B. Tuning the electrical properties of the heart by differential trafficking of KATP ion channel complexes. J Cell Sci 2014; 127:2106-19. [PMID: 24569881 PMCID: PMC4004980 DOI: 10.1242/jcs.141440] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The copy number of membrane proteins at the cell surface is tightly regulated. Many ion channels and receptors present retrieval motifs to COPI vesicle coats and are retained in the early secretory pathway. In some cases, the interaction with COPI is prevented by binding to 14-3-3 proteins. However, the functional significance of this antagonism between COPI and 14-3-3 in terminally differentiated cells is unknown. Here, we show that ATP-sensitive K+ (KATP) channels, which are composed of Kir6.2 and SUR1 subunits, are stalled in the Golgi complex of ventricular, but not atrial, cardiomyocytes. Upon sustained β-adrenergic stimulation, which leads to activation of protein kinase A (PKA), SUR1-containing channels reach the plasma membrane of ventricular cells. We show that PKA-dependent phosphorylation of the C-terminus of Kir6.2 decreases binding to COPI and, thereby, silences the arginine-based retrieval signal. Thus, activation of the sympathetic nervous system releases this population of KATP channels from storage in the Golgi and, hence, might facilitate the adaptive response to metabolic challenges.
Collapse
Affiliation(s)
- Eric C Arakel
- Department of Molecular Biology, Center for Biochemistry and Molecular Cell Biology, Heart Research Center Göttingen, University Medicine Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Renigunta V, Zou X, Kling S, Schlichthörl G, Daut J. Breaking the silence: functional expression of the two-pore-domain potassium channel THIK-2. Pflugers Arch 2013; 466:1735-45. [DOI: 10.1007/s00424-013-1404-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 11/09/2013] [Indexed: 10/26/2022]
|
39
|
|
40
|
A splice variant of the two-pore domain potassium channel TREK-1 with only one pore domain reduces the surface expression of full-length TREK-1 channels. Pflugers Arch 2013; 466:1559-70. [DOI: 10.1007/s00424-013-1384-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/09/2013] [Accepted: 10/12/2013] [Indexed: 10/26/2022]
|
41
|
Chatelain FC, Bichet D, Feliciangeli S, Larroque MM, Braud VM, Douguet D, Lesage F. Silencing of the tandem pore domain halothane-inhibited K+ channel 2 (THIK2) relies on combined intracellular retention and low intrinsic activity at the plasma membrane. J Biol Chem 2013; 288:35081-92. [PMID: 24163367 DOI: 10.1074/jbc.m113.503318] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The tandem pore domain halothane-inhibited K(+) channel 1 (THIK1) produces background K(+) currents. Despite 62% amino acid identity with THIK1, THIK2 is not active upon heterologous expression. Here, we show that this apparent lack of activity is due to a unique combination of retention in the endoplasmic reticulum and low intrinsic channel activity at the plasma membrane. A THIK2 mutant containing a proline residue (THIK2-A155P) in its second inner helix (M2) produces K(+)-selective currents with properties similar to THIK1, including inhibition by halothane and insensitivity to extracellular pH variations. Another mutation in the M2 helix (I158D) further increases channel activity and affects current kinetics. We also show that the cytoplasmic amino-terminal region of THIK2 (Nt-THIK2) contains an arginine-rich motif (RRSRRR) that acts as a retention/retrieval signal. Mutation of this motif in THIK2 induces a relocation of the channel to the plasma membrane, resulting in measurable currents, even in the absence of mutations in the M2 helix. Cell surface delivery of a Nt-THIK2-CD161 chimera is increased by mutating the arginines of the retention motif but also by converting the serine embedded in this motif to aspartate, suggesting a phosphorylation-dependent regulation of THIK2 trafficking.
Collapse
Affiliation(s)
- Franck C Chatelain
- From the Laboratory of Excellence Ion Channel Science and Therapeutics, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, and Université de Nice Sophia Antipolis, 660 Route des Lucioles, 06560 Valbonne, France
| | | | | | | | | | | | | |
Collapse
|
42
|
Brown M, Stafford LJ, Onisk D, Joaquim T, Tobb A, Goldman L, Fancy D, Stave J, Chambers R. Snorkel: an epitope tagging system for measuring the surface expression of membrane proteins. PLoS One 2013; 8:e73255. [PMID: 24023844 PMCID: PMC3759426 DOI: 10.1371/journal.pone.0073255] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/17/2013] [Indexed: 02/01/2023] Open
Abstract
Tags are widely used to monitor a protein’s expression level, interactions, protein trafficking, and localization. Membrane proteins are often tagged in their extracellular domains to allow discrimination between protein in the plasma membrane from that in internal pools. Multipass membrane proteins offer special challenges for inserting a tag since the extracellular regions are often composed of small loops and thus inserting an epitope tag risks perturbing the structure, function, or location of the membrane protein. We have developed a novel tagging system called snorkel where a transmembrane domain followed by a tag is appended to the cytoplasmic C-terminus of the membrane protein. In this way the tag is displayed extracellularly, but structurally separate from the membrane protein. We have tested the snorkel tag system on a diverse panel of membrane proteins including GPCRs and ion channels and demonstrated that it reliably allows for monitoring of the surface expression.
Collapse
Affiliation(s)
| | | | - Dale Onisk
- SDIX, Newark, Delaware, United States of America
| | - Tony Joaquim
- SDIX, Newark, Delaware, United States of America
| | - Alhagie Tobb
- SDIX, Newark, Delaware, United States of America
| | | | - David Fancy
- SDIX, Newark, Delaware, United States of America
| | - James Stave
- SDIX, Newark, Delaware, United States of America
| | - Ross Chambers
- SDIX, Newark, Delaware, United States of America
- * E-mail:
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Potassium channels in the distal nephron are precisely controlled to regulate potassium secretion in accord with physiological demands. In recent years, it has become evident that membrane trafficking processes play a fundamental role. This short review highlights recent developments in elucidating the underlying mechanisms. RECENT FINDINGS Novel sorting signals in the renal potassium channels, and the elusive intracellular trafficking machinery that read and act on these signals have recently been identified. These new discoveries reveal that independent signals sequentially interact with different intracellular sorting, retention and internalization machineries to appropriately ferry the channels to and from the apical and basolateral membrane domains in sufficient numbers to regulate potassium balance. SUMMARY A new understanding of the basic mechanisms that control potassium channel density at polarized membrane domains has emerged, providing new insights into how potassium balance is achieved and how it goes awry in disease.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland Medical School, Baltimore, Maryland 21201, USA.
| |
Collapse
|
44
|
Ma L, Roman-Campos D, Austin ED, Eyries M, Sampson KS, Soubrier F, Germain M, Trégouët DA, Borczuk A, Rosenzweig EB, Girerd B, Montani D, Humbert M, Loyd JE, Kass RS, Chung WK. A novel channelopathy in pulmonary arterial hypertension. N Engl J Med 2013; 369:351-361. [PMID: 23883380 PMCID: PMC3792227 DOI: 10.1056/nejmoa1211097] [Citation(s) in RCA: 337] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension is a devastating disease with high mortality. Familial cases of pulmonary arterial hypertension are usually characterized by autosomal dominant transmission with reduced penetrance, and some familial cases have unknown genetic causes. METHODS We studied a family in which multiple members had pulmonary arterial hypertension without identifiable mutations in any of the genes known to be associated with the disease, including BMPR2, ALK1, ENG, SMAD9, and CAV1. Three family members were studied with whole-exome sequencing. Additional patients with familial or idiopathic pulmonary arterial hypertension were screened for the mutations in the gene that was identified on whole-exome sequencing. All variants were expressed in COS-7 cells, and channel function was studied by means of patch-clamp analysis. RESULTS We identified a novel heterozygous missense variant c.608 G→A (G203D) in KCNK3 (the gene encoding potassium channel subfamily K, member 3) as a disease-causing candidate gene in the family. Five additional heterozygous missense variants in KCNK3 were independently identified in 92 unrelated patients with familial pulmonary arterial hypertension and 230 patients with idiopathic pulmonary arterial hypertension. We used in silico bioinformatic tools to predict that all six novel variants would be damaging. Electrophysiological studies of the channel indicated that all these missense mutations resulted in loss of function, and the reduction in the potassium-channel current was remedied by the application of the phospholipase inhibitor ONO-RS-082. CONCLUSIONS Our study identified the association of a novel gene, KCNK3, with familial and idiopathic pulmonary arterial hypertension. Mutations in this gene produced reduced potassium-channel current, which was successfully remedied by pharmacologic manipulation. (Funded by the National Institutes of Health.)
Collapse
Affiliation(s)
- Lijiang Ma
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Danilo Roman-Campos
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Eric D Austin
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Mélanie Eyries
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Kevin S Sampson
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Florent Soubrier
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Marine Germain
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - David-Alexandre Trégouët
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Alain Borczuk
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Erika Berman Rosenzweig
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Barbara Girerd
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - David Montani
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Marc Humbert
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - James E Loyd
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Robert S Kass
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| | - Wendy K Chung
- Departments of Pediatrics (L.M., E.B.R., W.K.C.), Pharmacology (D.R.-C., K.S.S., R.S.K.), and Pathology (A.B.), Columbia University Medical Center, New York; the Departments of Pediatrics (E.D.A.) and Medicine (J.E.L.), Vanderbilt University Medical Center, Nashville; the Genetics Department, Hospital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Institut National de la Santé et de la Recherche Médicale (INSERM), and Université Pierre et Marie Curie (UPMC) Unité Mixte de Recherche en Santé (UMRS) 956, Institute of Cardiometabolism and Nutrition (ICAN) (M.E., F.S.); and INSERM-UPMC UMRS 937, ICAN (M.G., D.-A.T.) - all in Paris; and APHP, Département Hospitalo-Universitaire Thorax Innovation (DHU TORINO), Service de Pneumologie, Hôpital Bicêtre; Université Paris-Sud, Laboratoire d'Excellence en Recherche sur le Médicament et Innovation Thérapeutique (LERMIT); and INSERM UMRS 999 - all in Le Kremlin-Bicêtre, France (B.G., D.M., M.H.)
| |
Collapse
|
45
|
Maddala R, Nagendran T, de Ridder GG, Schey KL, Rao PV. L-type calcium channels play a critical role in maintaining lens transparency by regulating phosphorylation of aquaporin-0 and myosin light chain and expression of connexins. PLoS One 2013; 8:e64676. [PMID: 23734214 PMCID: PMC3667166 DOI: 10.1371/journal.pone.0064676] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/17/2013] [Indexed: 01/18/2023] Open
Abstract
Homeostasis of intracellular calcium is crucial for lens cytoarchitecture and transparency, however, the identity of specific channel proteins regulating calcium influx within the lens is not completely understood. Here we examined the expression and distribution profiles of L-type calcium channels (LTCCs) and explored their role in morphological integrity and transparency of the mouse lens, using cDNA microarray, RT-PCR, immunoblot, pharmacological inhibitors and immunofluorescence analyses. The results revealed that Ca (V) 1.2 and 1.3 channels are expressed and distributed in both the epithelium and cortical fiber cells in mouse lens. Inhibition of LTCCs with felodipine or nifedipine induces progressive cortical cataract formation with time, in association with decreased lens weight in ex-vivo mouse lenses. Histological analyses of felodipine treated lenses revealed extensive disorganization and swelling of cortical fiber cells resembling the phenotype reported for altered aquaporin-0 activity without detectable cytotoxic effects. Analysis of both soluble and membrane rich fractions from felodipine treated lenses by SDS-PAGE in conjunction with mass spectrometry and immunoblot analyses revealed decreases in β-B1-crystallin, Hsp-90, spectrin and filensin. Significantly, loss of transparency in the felodipine treated lenses was preceded by an increase in aquaporin-0 serine-235 phosphorylation and levels of connexin-50, together with decreases in myosin light chain phosphorylation and the levels of 14-3-3ε, a phosphoprotein-binding regulatory protein. Felodipine treatment led to a significant increase in gene expression of connexin-50 and 46 in the mouse lens. Additionally, felodipine inhibition of LTCCs in primary cultures of mouse lens epithelial cells resulted in decreased intracellular calcium, and decreased actin stress fibers and myosin light chain phosphorylation, without detectable cytotoxic response. Taken together, these observations reveal a crucial role for LTCCs in regulation of expression, activity and stability of aquaporin-0, connexins, cytoskeletal proteins, and the mechanical properties of lens, all of which have a vital role in maintaining lens function and cytoarchitecture.
Collapse
Affiliation(s)
- Rupalatha Maddala
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Tharkika Nagendran
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Gustaaf G. de Ridder
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Ponugoti Vasantha Rao
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
46
|
Anders C, Higuchi Y, Koschinsky K, Bartel M, Schumacher B, Thiel P, Nitta H, Preisig-Müller R, Schlichthörl G, Renigunta V, Ohkanda J, Daut J, Kato N, Ottmann C. A Semisynthetic Fusicoccane Stabilizes a Protein-Protein Interaction and Enhances the Expression of K+ Channels at the Cell Surface. ACTA ACUST UNITED AC 2013; 20:583-93. [DOI: 10.1016/j.chembiol.2013.03.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 03/21/2013] [Accepted: 03/25/2013] [Indexed: 01/01/2023]
|
47
|
Li JG, Chen C, Huang P, Wang Y, Liu-Chen LY. 14-3-3ζ Protein regulates anterograde transport of the human κ-opioid receptor (hKOPR). J Biol Chem 2012; 287:37778-92. [PMID: 22989890 DOI: 10.1074/jbc.m112.359679] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
By proteomic analysis, we found that 14-3-3ζ was one of the proteins co-immunoprecipitated with human κ-opioid receptor (hKOPR) from extracts of solubilized Neuro2A cells stably expressing FLAG-hKOPR (N2A-FLAG-hKOPR cells). 14-3-3 proteins are a family of conserved regulatory molecules in eukaryotic cells, where they participate in signal transduction, metabolism, and membrane protein transport. 14-3-3ζ co-localized with the hKOPR in N2A cells. The hKOPR C-tail interacted with 14-3-3ζ in rat brain extracts and bound directly to purified 14-3-3ζ as demonstrated by pulldown techniques. 14-3-3ζ siRNA decreased expression of the hKOPR in N2A-FLAG-hKOPR cells and cultured primary cortical neurons of E19 rats by ~25% as determined by immunoblotting, ligand binding, and flow cytometry. The effect of 14-3-3ζ siRNA was reversed by overexpression of 14-3-3ζ. Expression of the 14-3-3 scavenger protein pGpLI-R18 also decreased hKOPR expression. 14-3-3ζ siRNA did not change expressions of the hDOPR and rMOPR in N2A cells. Pulse-chase study showed that 14-3-3ζ siRNA decreased the amount of mature hKOPR but did not change the rate of maturation or stability of hKOPR protein. Mutations of R354A/S358A in the putative 14-3-3 interaction motif (354)RQSTS(358) in the hKOPR C-tail reduced interaction of the hKOPR with 14-3-3ζ and abolished the effect of 14-3-3ζ knockdown on hKOPR expression. Mutation of the endoplasmic reticulum retention motif (359)RVR adjacent to the 14-3-3 interaction motif in the hKOPR C-tail decreased interaction of coatomer protein I (COPI) with the hKOPR and abolished 14-3-3ζ-mediated regulation of hKOPR expression. 14-3-3ζ knockdown increased association of COPI with the hKOPR. These results suggest that 14-3-3ζ promotes expression of the hKOPR by inhibiting COPI and RVR motif-mediated endoplasmic reticulum localization machinery.
Collapse
Affiliation(s)
- Jian-Guo Li
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | |
Collapse
|
48
|
Schiekel J, Lindner M, Hetzel A, Wemhöner K, Renigunta V, Schlichthörl G, Decher N, Oliver D, Daut J. The inhibition of the potassium channel TASK-1 in rat cardiac muscle by endothelin-1 is mediated by phospholipase C. Cardiovasc Res 2012; 97:97-105. [DOI: 10.1093/cvr/cvs285] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
49
|
Gabriel L, Lvov A, Orthodoxou D, Rittenhouse AR, Kobertz WR, Melikian HE. The acid-sensitive, anesthetic-activated potassium leak channel, KCNK3, is regulated by 14-3-3β-dependent, protein kinase C (PKC)-mediated endocytic trafficking. J Biol Chem 2012; 287:32354-66. [PMID: 22846993 DOI: 10.1074/jbc.m112.391458] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The acid-sensitive neuronal potassium leak channel, KCNK3, is vital for setting the resting membrane potential and is the primary target for volatile anesthetics. Recent reports demonstrate that KCNK3 activity is down-regulated by PKC; however, the mechanisms responsible for PKC-induced KCNK3 down-regulation are undefined. Here, we report that endocytic trafficking dynamically regulates KCNK3 activity. Phorbol esters and Group I metabotropic glutamate receptor (mGluR) activation acutely decreased both native and recombinant KCNK3 currents with concomitant KCNK3 surface losses in cerebellar granule neurons and cell lines. PKC-mediated KCNK3 internalization required the presence of both 14-3-3β and a novel potassium channel endocytic motif, because depleting either 14-3-3β protein levels or ablating the endocytic motif completely abrogated PKC-regulated KCNK3 trafficking. These results demonstrate that neuronal potassium leak channels are not static membrane residents but are subject to 14-3-3β-dependent regulated trafficking, providing a straightforward mechanism to modulate neuronal excitability and synaptic plasticity by Group I mGluRs.
Collapse
Affiliation(s)
- Luke Gabriel
- Graduate Program in Neuroscience, University of Massachusetts Medical School, Worcester, Massachusetts 01604, USA
| | | | | | | | | | | |
Collapse
|
50
|
O'Brien JE, Sharkey LM, Vallianatos CN, Han C, Blossom JC, Yu T, Waxman SG, Dib-Hajj SD, Meisler MH. Interaction of voltage-gated sodium channel Nav1.6 (SCN8A) with microtubule-associated protein Map1b. J Biol Chem 2012; 287:18459-66. [PMID: 22474336 DOI: 10.1074/jbc.m111.336024] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mechanism by which voltage-gated sodium channels are trafficked to the surface of neurons is not well understood. Our previous work implicated the cytoplasmic N terminus of the sodium channel Na(v)1.6 in this process. We report that the N terminus plus the first transmembrane segment (residues 1-153) is sufficient to direct a reporter to the cell surface. To identify proteins that interact with the 117-residue N-terminal domain, we carried out a yeast two-hybrid screen of a mouse brain cDNA library. Three clones containing overlapping portions of the light chain of microtubule-associated protein Map1b (Mtap1b) were recovered from the screen. Interaction between endogenous Na(v)1.6 channels and Map1b in mouse brain was confirmed by co-immunoprecipitation. Map1b did not interact with the N terminus of the related channel Na(v)1.1. Alanine-scanning mutagenesis of the Na(v)1.6 N terminus demonstrated that residues 77-80 (VAVP) contribute to interaction with Map1b. Co-expression of Na(v)1.6 with Map1b in neuronal cell line ND7/23 resulted in a 50% increase in current density, demonstrating a functional role for this interaction. Mutation of the Map1b binding site of Na(v)1.6 prevented generation of sodium current in transfected cells. The data indicate that Map1b facilitates trafficking of Na(v)1.6 to the neuronal cell surface.
Collapse
Affiliation(s)
- Janelle E O'Brien
- Department of Human Genetics, the University of Michigan, Ann Arbor, Michigan 48109-5618, USA
| | | | | | | | | | | | | | | | | |
Collapse
|