1
|
Rajeev P, Singh N, Kechkar A, Butler C, Ramanan N, Sibarita JB, Jose M, Nair D. Nanoscale regulation of Ca2+ dependent phase transitions and real-time dynamics of SAP97/hDLG. Nat Commun 2022; 13:4236. [PMID: 35869063 PMCID: PMC9307800 DOI: 10.1038/s41467-022-31912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Synapse associated protein-97/Human Disk Large (SAP97/hDLG) is a conserved, alternatively spliced, modular, scaffolding protein critical in regulating the molecular organization of cell-cell junctions in vertebrates. We confirm that the molecular determinants of first order phase transition of SAP97/hDLG is controlled by morpho-functional changes in its nanoscale organization. Furthermore, the nanoscale molecular signatures of these signalling islands and phase transitions are altered in response to changes in cytosolic Ca2+. Additionally, exchange kinetics of alternatively spliced isoforms of the intrinsically disordered region in SAP97/hDLG C-terminus shows differential sensitivities to Ca2+ bound Calmodulin, affirming that the molecular signatures of local phase transitions of SAP97/hDLG depends on their nanoscale heterogeneity and compositionality of isoforms. SAP97/hDLG is a ubiquitous, alternatively spliced, and conserved modular scaffolding protein involved in the organization cell junctions and excitatory synapses. Here, authors confirm that SAP97/hDLG condenses in to nanosized molecular domains in both heterologous cells and hippocampal pyramidal neurons. Authors demonstrate that in vivo and in vitro condensation, molecular signatures of nanoscale condensates and exchange kinetics of SAP97/hDLG is modulated by the local availability of alternatively spliced isoforms. Additionally, SAP97/hDLG isoforms exhibits a differential sensitivity to Ca2+ bound Calmodulin, resulting in altered properties of nanocondensates and their real-time regulation
Collapse
|
2
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
3
|
Lewin L, Nsasra E, Golbary E, Hadad U, Orr I, Yifrach O. Molecular and cellular correlates in Kv channel clustering: entropy-based regulation of cluster ion channel density. Sci Rep 2020; 10:11304. [PMID: 32647278 PMCID: PMC7347538 DOI: 10.1038/s41598-020-68003-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/27/2020] [Indexed: 11/25/2022] Open
Abstract
Scaffold protein-mediated ion channel clustering at unique membrane sites is important for electrical signaling. Yet, the mechanism(s) by which scaffold protein-ion channel interactions lead to channel clustering or how cluster ion channel density is regulated is mostly not known. The voltage-activated potassium channel (Kv) represents an excellent model to address these questions as the mechanism underlying its interaction with the post-synaptic density 95 (PSD-95) scaffold protein is known to be controlled by the length of the extended ‘ball and chain’ sequence comprising the C-terminal channel region. Here, using sub-diffraction high-resolution imaging microscopy, we show that Kv channel ‘chain’ length regulates Kv channel density with a ‘bell’-shaped dependence, reflecting a balance between thermodynamic considerations controlling ‘chain’ recruitment by PSD-95 and steric hindrance due to the spatial proximity of multiple channel molecules. Our results thus reveal an entropy-based mode of channel cluster density regulation that mirrors the entropy-based regulation of the Kv channel-PSD-95 interaction. The implications of these findings for electrical signaling are discussed.
Collapse
Affiliation(s)
- Limor Lewin
- Department of Life Sciences and the Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Esraa Nsasra
- Department of Life Sciences and the Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Ella Golbary
- Department of Life Sciences and the Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Uzi Hadad
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Irit Orr
- Department of Life Sciences and the Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Ofer Yifrach
- Department of Life Sciences and the Zlotowski Center for Neurosciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| |
Collapse
|
4
|
Ma R, Lewis A. Spadin Selectively Antagonizes Arachidonic Acid Activation of TREK-1 Channels. Front Pharmacol 2020; 11:434. [PMID: 32317978 PMCID: PMC7154116 DOI: 10.3389/fphar.2020.00434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
TREK-1 channel activity is a critical regulator of neuronal, cardiac, and smooth muscle physiology and pathology. The antidepressant peptide, spadin, has been proposed to be a TREK-1-specific blocker. Here we sought to examine the mechanism of action underlying spadin inhibition of TREK-1 channels. Heterologous expression in Xenopus laevis oocytes and electrophysiological analysis using two-electrode voltage clamp in standard bath solutions was used to characterize the pharmacological profile of wild-type and mutant murine TREK-1 and TREK-2 channels using previously established human K2P activators; arachidonic acid (AA), cis-4,7,10,13,16,19-docosahexaenoic acid (DHA), BL-1249, and cinnamyl-3,4-dihydroxy-α-cyanocinnamate (CDC) and inhibitors; spadin and barium (Ba2+). Mouse TREK-1 and TREK-2 channel currents were both significantly increased by AA, BL-1249, and CDC, similar to their human homologs. Under basal conditions, both TREK-1 and TREK-2 currents were insensitive to application of spadin, but could be blocked by Ba2+. Spadin did not significantly inhibit either TREK-1 or TREK-2 currents either chemically activated by AA, BL-1249, or CDC, or structurally activated via a gating mutation. However, pre-exposure to spadin significantly perturbed the subsequent activation of TREK-1 currents by AA, but not TREK-2. Furthermore, spadin was unable to prevent activation of TREK-1 by BL-1249, CDC, or the related bioactive lipid, DHA. Spadin specifically antagonizes the activation of TREK-1 channels by AA, likely via an allosteric mechanism. Lack of intrinsic activity may explain the absence of clinical side effects during antidepressant therapy.
Collapse
Affiliation(s)
- Ruolin Ma
- School of Pharmacy and Biomedical Sciences, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Anthony Lewis
- School of Pharmacy and Biomedical Sciences, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
5
|
Translating preclinical findings in clinically relevant new antipsychotic targets: focus on the glutamatergic postsynaptic density. Implications for treatment resistant schizophrenia. Neurosci Biobehav Rev 2019; 107:795-827. [DOI: 10.1016/j.neubiorev.2019.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/20/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
|
6
|
Siddiqui S, Khan F, Jamali KS, Musharraf SG. Madecassic Acid Reduces Fast Transient Potassium Channels and Promotes Neurite Elongation in Hippocampal CA1 Neurons. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 19:12-26. [PMID: 31713492 DOI: 10.2174/1871527318666191111105508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVE Madecassic Acid (MA) is well known to induce neurite elongation. However, its correlation with the expression of fast transient potassium (AKv) channels during neuronal development has not been well studied. Therefore, the present study was designed to investigate the effects of MA on the modulation of AKv channels during neurite outgrowth. METHODS Neurite outgrowth was measured with morphometry software, and Kv4 currents were recorded by using the patch clamp technique. RESULTS The ability of MA to promote neurite outgrowth is dose-dependent and was blocked by using the mitogen/extracellular signal-regulated kinase (MEK) inhibitor U0126. MA reduced the peak current density and surface expression of the AKv channel Kv4.2 with or without the presence of NaN3. The surface expression of Kv4.2 channels was also reduced after MA treatment of growing neurons. Ethylene glycol tetraacetic acid (EGTA) and an N-methyl-D-aspartate (NMDA) receptor blocker, MK801 along with MA prevented the effect of MA on neurite length, indicating that calcium entry through NMDA receptors is necessary for MA-induced neurite outgrowth. CONCLUSION The data demonstrated that MA increased neurite outgrowth by internalizing AKv channels in neurons. Any alterations in the precise density of ion channels can lead to deleterious consequences on health because it changes the electrical and mechanical function of a neuron or a cell. Modulating ion channel's density is exciting research in order to develop novel drugs for the therapeutic treatment of various diseases of CNS.
Collapse
Affiliation(s)
- Sonia Siddiqui
- Department of Biochemistry, Dow University of Health Sciences (DUHS), Karachi, Pakistan.,Department of Neuroscience, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi 75270, Pakistan
| | - Faisal Khan
- Department of Neuroscience, Dr. Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences (ICCBS), University of Karachi 75270, Pakistan
| | - Khawar Saeed Jamali
- Department of Surgery, Dow University of Health Sciences (DUHS), Karachi, Pakistan
| | - Syed Ghulam Musharraf
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi 75270, Pakistan
| |
Collapse
|
7
|
Hyperexcitability and Hyperplasticity Disrupt Cerebellar Signal Transfer in the IB2 KO Mouse Model of Autism. J Neurosci 2019; 39:2383-2397. [PMID: 30696733 DOI: 10.1523/jneurosci.1985-18.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/22/2018] [Accepted: 01/08/2019] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorders (ASDs) are pervasive neurodevelopmental conditions that often involve mutations affecting synaptic mechanisms. Recently, the involvement of cerebellum in ASDs has been suggested, but the underlying functional alterations remained obscure. We investigated single-neuron and microcircuit properties in IB2 (Islet Brain-2) KO mice of either sex. The IB2 gene (chr22q13.3 terminal region) deletion occurs in virtually all cases of Phelan-McDermid syndrome, causing autistic symptoms and a severe delay in motor skill acquisition. IB2 KO granule cells showed a larger NMDA receptor-mediated current and enhanced intrinsic excitability, raising the excitatory/inhibitory balance. Furthermore, the spatial organization of granular layer responses to mossy fibers shifted from a "Mexican hat" to a "stovepipe hat" profile, with stronger excitation in the core and weaker inhibition in the surround. Finally, the size and extension of long-term synaptic plasticity were remarkably increased. These results show for the first time that hyperexcitability and hyperplasticity disrupt signal transfer in the granular layer of IB2 KO mice, supporting cerebellar involvement in the pathogenesis of ASD.SIGNIFICANCE STATEMENT This article shows for the first time a complex set of alterations in the cerebellum granular layer of a mouse model [IB2 (Islet Brain-2) KO] of autism spectrum disorders. The IB2 KO in mice mimics the deletion of the corresponding gene in the Phelan-McDermid syndrome in humans. The changes reported here are centered on NMDA receptor hyperactivity, hyperplasticity, and hyperexcitability. These, in turn, increase the excitatory/inhibitory balance and alter the shape of center/surround structures that emerge in the granular layer in response to mossy fiber activity. These results support recent theories suggesting the involvement of cerebellum in autism spectrum disorders.
Collapse
|
8
|
Rhee SW, Rusch NJ. Molecular determinants of beta-adrenergic signaling to voltage-gated K + channels in the cerebral circulation. Microcirculation 2018; 25. [PMID: 29072364 DOI: 10.1111/micc.12425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022]
Abstract
Voltage-gated K+ (Kv ) channels are major determinants of membrane potential in vascular smooth muscle cells (VSMCs) and regulate the diameter of small cerebral arteries and arterioles. However, the intracellular structures that govern the expression and function of vascular Kv channels are poorly understood. Scaffolding proteins including postsynaptic density 95 (PSD95) recently were identified in rat cerebral VSMCs. Primarily characterized in neurons, the PSD95 scaffold has more than 50 known binding partners, and it can mediate macromolecular signaling between cell-surface receptors and ion channels. In cerebral arteries, Shaker-type Kv 1 channels appear to associate with the PSD95 molecular scaffold, and PSD95 is required for the normal expression and vasodilator influence of members of this K+ channel gene family. Furthermore, recent findings suggest that the β1-subtype adrenergic receptor is expressed in cerebral VSMCs and forms a functional vasodilator complex with Kv 1 channels on the PSD95 scaffold. Activation of β1-subtype adrenergic receptors in VSMCs enables protein kinase A-dependent phosphorylation and opening of Kv 1 channels in the PSD95 complex; the subsequent K+ efflux mediates membrane hyperpolarization and vasodilation of small cerebral arteries. Early evidence from other studies suggests that other families of Kv channels and scaffolding proteins are expressed in VSMCs. Future investigations into these macromolecular complexes that modulate the expression and function of Kv channels may reveal unknown signaling cascades that regulate VSMC excitability and provide novel targets for ion channel-based medications to optimize vascular tone.
Collapse
Affiliation(s)
- Sung W Rhee
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
9
|
Abstract
Activation of the electrical signal and its transmission as a depolarizing wave in the whole heart requires highly organized myocyte architecture and cell-cell contacts. In addition, complex trafficking and anchoring intracellular machineries regulate the proper surface expression of channels and their targeting to distinct membrane domains. An increasing list of proteins, lipids, and second messengers can contribute to the normal targeting of ion channels in cardiac myocytes. However, their precise roles in the electrophysiology of the heart are far from been extensively understood. Nowadays, much effort in the field focuses on understanding the mechanisms that regulate ion channel targeting to sarcolemma microdomains and their organization into macromolecular complexes. The purpose of the present section is to provide an overview of the characterized partners of the main cardiac sodium channel, NaV1.5, involved in regulating the functional expression of this channel both in terms of trafficking and targeting into microdomains.
Collapse
|
10
|
Lively S, Lam D, Wong R, Schlichter LC. Comparing Effects of Transforming Growth Factor β1 on Microglia From Rat and Mouse: Transcriptional Profiles and Potassium Channels. Front Cell Neurosci 2018; 12:115. [PMID: 29780305 PMCID: PMC5946019 DOI: 10.3389/fncel.2018.00115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/11/2018] [Indexed: 12/02/2022] Open
Abstract
The cytokine, transforming growth factor β1 (TGFβ1), is up-regulated after central nervous system (CNS) injuries or diseases involving microglial activation, and it has been proposed as a therapeutic agent for treating neuroinflammation. Microglia can produce and respond to TGFβ1. While rats and mice are commonly used for studying neuroinflammation, very few reports directly compare them. Such studies are important for improving pre-clinical studies and furthering translational progress in developing therapeutic interventions. After intracerebral hemorrhage (ICH) in the rat striatum, the TGFβ1 receptor was highly expressed on microglia/macrophages within the hematoma. We recently found species similarities and differences in response to either a pro-inflammatory (interferon-γ, IFN-γ, +tumor necrosis factor, TNF-α) or anti-inflammatory interleukin-4 (IL-4) stimulus. Here, we assessed whether rat and mouse microglia differ in their responses to TGFβ1. Microglia were isolated from Sprague-Dawley rats and C57BL/6 mice and treated with TGFβ1. We quantified changes in expression of >50 genes, in their morphology, proliferation, apoptosis and in three potassium channels that are considered therapeutic targets. Many inflammatory mediators, immune receptors and modulators showed species similarities, but notable differences included that, for some genes, only one species responded (e.g., Il4r, Il10, Tgfbr2, colony-stimulating factor receptor (Csf1r), Itgam, suppressor of cytokine signaling 1 (Socs1), toll-like receptors 4 (Tlr4), P2rx7, P2ry12), and opposite responses were seen for others (Tgfb1, Myc, Ifngr1). In rat only, TGFβ1 affected microglial morphology and proliferation, but there was no apoptosis in either species. In both species, TGFβ1 dramatically increased Kv1.3 channel expression and current (no effects on Kir2.1). KCa3.1 showed opposite species responses: the current was low in unstimulated rat microglia and greatly increased by TGFβ1 but higher in control mouse cells and decreased by TGFβ1. Finally, we compared TGFβ1 and IL10 (often considered similar anti-inflammatory stimuli) and found many different responses in both species. Overall, the numerous species differences should be considered when characterizing neuroinflammation and microglial activation in vitro and in vivo, and when targeting potassium channels.
Collapse
Affiliation(s)
- Starlee Lively
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada
| | - Doris Lam
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Raymond Wong
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Lyanne C Schlichter
- Krembil Research Institute, Genes and Development Division, University Health Network, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Voros O, Szilagyi O, Balajthy A, Somodi S, Panyi G, Hajdu P. The C-terminal HRET sequence of Kv1.3 regulates gating rather than targeting of Kv1.3 to the plasma membrane. Sci Rep 2018; 8:5937. [PMID: 29650988 PMCID: PMC5897520 DOI: 10.1038/s41598-018-24159-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/08/2018] [Indexed: 12/13/2022] Open
Abstract
Kv1.3 channels are expressed in several cell types including immune cells, such as T lymphocytes. The targeting of Kv1.3 to the plasma membrane is essential for T cell clonal expansion and assumed to be guided by the C-terminus of the channel. Using two point mutants of Kv1.3 with remarkably different features compared to the wild-type Kv1.3 (A413V and H399K having fast inactivation kinetics and tetraethylammonium-insensitivity, respectively) we showed that both Kv1.3 channel variants target to the membrane when the C-terminus was truncated right after the conserved HRET sequence and produce currents identical to those with a full-length C-terminus. The truncation before the HRET sequence (NOHRET channels) resulted in reduced membrane-targeting but non-functional phenotypes. NOHRET channels did not display gating currents, and coexpression with wild-type Kv1.3 did not rescue the NOHRET-A413V phenotype, no heteromeric current was observed. Interestingly, mutants of wild-type Kv1.3 lacking HRET(E) (deletion) or substituted with five alanines for the HRET(E) motif expressed current indistinguishable from the wild-type. These results demonstrate that the C-terminal region of Kv1.3 immediately proximal to the S6 helix is required for the activation gating and conduction, whereas the presence of the distal region of the C-terminus is not exclusively required for trafficking of Kv1.3 to the plasma membrane.
Collapse
Affiliation(s)
- Orsolya Voros
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary
| | - Orsolya Szilagyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary
| | - András Balajthy
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary
| | - Sándor Somodi
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 1 Egyetem sq., 4032, Hungary. MTA-DE-NAP B Ion Channel Structure-Function Research Group, RCMM, University of Debrecen, 400, Debrecen, Hungary
| | - Péter Hajdu
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary. .,Department of Biophysics and Cell Biology, Faculty of Dentistry, University of Debrecen, 400, 1 Egyetem sq., Debrecen, 4032, Hungary.
| |
Collapse
|
12
|
Kurokawa T, Kiyonaka S, Nakata E, Endo M, Koyama S, Mori E, Tran NH, Dinh H, Suzuki Y, Hidaka K, Kawata M, Sato C, Sugiyama H, Morii T, Mori Y. DNA Origami Scaffolds as Templates for Functional Tetrameric Kir3 K +
Channels. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201709982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tatsuki Kurokawa
- Department of Synthetic Chemistry and Biological Chemistry; Graduate School of Engineering; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry; Graduate School of Engineering; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Department of Technology and Ecology, Hall of Global Environmental Studies; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
| | - Eiji Nakata
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Institute of Advanced Energy; Kyoto University, Gokasho, Uji; Kyoto 611-0011 Japan
| | - Masayuki Endo
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku; Kyoto 606-8501 Japan
| | - Shohei Koyama
- Department of Synthetic Chemistry and Biological Chemistry; Graduate School of Engineering; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
| | - Emiko Mori
- Department of Synthetic Chemistry and Biological Chemistry; Graduate School of Engineering; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
| | - Nam Ha Tran
- Department of Technology and Ecology, Hall of Global Environmental Studies; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
| | - Huyen Dinh
- Institute of Advanced Energy; Kyoto University, Gokasho, Uji; Kyoto 611-0011 Japan
| | - Yuki Suzuki
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Department of Chemistry; Graduate School of Science; Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku; Kyoto 606-8502 Japan
| | - Kumi Hidaka
- Department of Chemistry; Graduate School of Science; Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku; Kyoto 606-8502 Japan
| | - Masaaki Kawata
- Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; 1-1-1 Higashi, Tsukuba Ibaraki 305-8566 Japan
| | - Chikara Sato
- Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; 1-1-1 Higashi, Tsukuba Ibaraki 305-8566 Japan
| | - Hiroshi Sugiyama
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku; Kyoto 606-8501 Japan
- Department of Chemistry; Graduate School of Science; Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku; Kyoto 606-8502 Japan
| | - Takashi Morii
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Institute of Advanced Energy; Kyoto University, Gokasho, Uji; Kyoto 611-0011 Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry; Graduate School of Engineering; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
- Core Research for Evolutional Science and Technology (Japan); Science and Technology Agency; 4-1-8 Hon-cho, Kawaguchi Saitama 332-0012 Japan
- Department of Technology and Ecology, Hall of Global Environmental Studies; Kyoto University, Kyotodaigakukatsura, Nishikyo-ku; Kyoto 615-8510 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku; Kyoto 606-8501 Japan
| |
Collapse
|
13
|
Kurokawa T, Kiyonaka S, Nakata E, Endo M, Koyama S, Mori E, Tran NH, Dinh H, Suzuki Y, Hidaka K, Kawata M, Sato C, Sugiyama H, Morii T, Mori Y. DNA Origami Scaffolds as Templates for Functional Tetrameric Kir3 K + Channels. Angew Chem Int Ed Engl 2018; 57:2586-2591. [PMID: 29341462 DOI: 10.1002/anie.201709982] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/15/2017] [Indexed: 01/17/2023]
Abstract
In native systems, scaffolding proteins play important roles in assembling proteins into complexes to transduce signals. This concept is yet to be applied to the assembly of functional transmembrane protein complexes in artificial systems. To address this issue, DNA origami has the potential to serve as scaffolds that arrange proteins at specific positions in complexes. Herein, we report that Kir3 K+ channel proteins are assembled through zinc-finger protein (ZFP)-adaptors at specific locations on DNA origami scaffolds. Specific binding of the ZFP-fused Kir3 channels and ZFP-based adaptors on DNA origami were confirmed by atomic force microscopy and gel electrophoresis. Furthermore, the DNA origami with ZFP binding sites nearly tripled the K+ channel current activity elicited by heterotetrameric Kir3 channels in HEK293T cells. Thus, our method provides a useful template to control the oligomerization states of membrane protein complexes in vitro and in living cells.
Collapse
Affiliation(s)
- Tatsuki Kurokawa
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan
| | - Shigeki Kiyonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Eiji Nakata
- Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Masayuki Endo
- Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shohei Koyama
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Emiko Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan
| | - Nam Ha Tran
- Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Huyen Dinh
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Yuki Suzuki
- Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Kumi Hidaka
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Masaaki Kawata
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Chikara Sato
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Hiroshi Sugiyama
- Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan.,Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Takashi Morii
- Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Core Research for Evolutional Science and Technology (Japan), Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama, 332-0012, Japan.,Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyotodaigakukatsura, Nishikyo-ku, Kyoto, 615-8510, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
14
|
|
15
|
Lo CA, Kays I, Emran F, Lin TJ, Cvetkovska V, Chen BE. Quantification of Protein Levels in Single Living Cells. Cell Rep 2015; 13:2634-2644. [PMID: 26686644 DOI: 10.1016/j.celrep.2015.11.048] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 09/16/2015] [Accepted: 11/13/2015] [Indexed: 10/22/2022] Open
Abstract
Accurate measurement of the amount of specific protein a cell produces is important for investigating basic molecular processes. We have developed a technique that allows for quantitation of protein levels in single cells in vivo. This protein quantitation ratioing (PQR) technique uses a genetic tag that produces a stoichiometric ratio of a fluorescent protein reporter and the protein of interest during protein translation. The fluorescence intensity is proportional to the number of molecules produced of the protein of interest and is used to determine the relative amount of protein within the cell. We use PQR to quantify protein expression of different genes using quantitative imaging, electrophysiology, and phenotype. We use genome editing to insert Protein Quantitation Reporters into endogenous genomic loci in three different genomes for quantitation of endogenous protein levels. The PQR technique will allow for a wide range of quantitative experiments examining gene-to-phenotype relationships with greater accuracy.
Collapse
Affiliation(s)
- Chiu-An Lo
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada
| | - Ibrahim Kays
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada
| | - Farida Emran
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada
| | - Tsung-Jung Lin
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada
| | - Vedrana Cvetkovska
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada
| | - Brian Edwin Chen
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, Montréal, QC H3G 1A4, Canada; Departments of Medicine and Neurology and Neurosurgery, McGill University, Montréal, QC H3G 1A4, Canada.
| |
Collapse
|
16
|
Alternative splicing modulates Kv channel clustering through a molecular ball and chain mechanism. Nat Commun 2015; 6:6488. [DOI: 10.1038/ncomms7488] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 02/02/2015] [Indexed: 01/22/2023] Open
|
17
|
Soudry D, Meir R. The neuronal response at extended timescales: a linearized spiking input-output relation. Front Comput Neurosci 2014; 8:29. [PMID: 24765073 PMCID: PMC3980113 DOI: 10.3389/fncom.2014.00029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/24/2014] [Indexed: 11/16/2022] Open
Abstract
Many biological systems are modulated by unknown slow processes. This can severely hinder analysis – especially in excitable neurons, which are highly non-linear and stochastic systems. We show the analysis simplifies considerably if the input matches the sparse “spiky” nature of the output. In this case, a linearized spiking Input–Output (I/O) relation can be derived semi-analytically, relating input spike trains to output spikes based on known biophysical properties. Using this I/O relation we obtain closed-form expressions for all second order statistics (input – internal state – output correlations and spectra), construct optimal linear estimators for the neuronal response and internal state and perform parameter identification. These results are guaranteed to hold, for a general stochastic biophysical neuron model, with only a few assumptions (mainly, timescale separation). We numerically test the resulting expressions for various models, and show that they hold well, even in cases where our assumptions fail to hold. In a companion paper we demonstrate how this approach enables us to fit a biophysical neuron model so it reproduces experimentally observed temporal firing statistics on days-long experiments.
Collapse
Affiliation(s)
- Daniel Soudry
- Laboratory for Network Biology Research, Department of Electrical Engineering Technion, Haifa, Israel
| | - Ron Meir
- Laboratory for Network Biology Research, Department of Electrical Engineering Technion, Haifa, Israel
| |
Collapse
|
18
|
Rickhag M, Hansen FH, Sørensen G, Strandfelt KN, Andresen B, Gotfryd K, Madsen KL, Vestergaard-Klewe I, Ammendrup-Johnsen I, Eriksen J, Newman AH, Füchtbauer EM, Gomeza J, Woldbye DPD, Wörtwein G, Gether U. A C-terminal PDZ domain-binding sequence is required for striatal distribution of the dopamine transporter. Nat Commun 2013; 4:1580. [PMID: 23481388 PMCID: PMC3646413 DOI: 10.1038/ncomms2568] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 01/31/2013] [Indexed: 01/27/2023] Open
Abstract
The dopamine transporter (DAT) mediates reuptake of dopamine from the synaptic cleft. The cellular mechanisms controlling DAT levels in striatal nerve terminals remain poorly understood. DAT contains a C-terminal PDZ (PSD-95/Discs-large/ZO-1) domain binding sequence believed to bind synaptic scaffolding proteins, but its functional significance is uncertain. Here we demonstrate that two different DAT knock-in mice with disrupted PDZ-binding motifs (DAT-AAA and DAT+Ala) are characterized by dramatic loss of DAT expression in the striatum, causing hyperlocomotion and attenuated response to amphetamine. In cultured dopaminergic neurons and striatal slices from DAT-AAA mice, we find markedly reduced DAT surface levels and evidence for enhanced constitutive internalization. In DAT-AAA neurons, but not in wild type neurons, surface levels are rescued in part by expression of a dominant-negative dynamin mutation (K44A). Our findings suggest that PDZ domain interactions are critical for synaptic distribution of DAT in vivo and thereby for proper maintenance of dopamine homeostasis.
Collapse
Affiliation(s)
- Mattias Rickhag
- Molecular Neuropharmacology Laboratory, Lundbeck Foundation Center for Biomembranes in Nanomedicine, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
The role of PSD-95 in the rearrangement of Kv1.3 channels to the immunological synapse. Pflugers Arch 2013; 465:1341-53. [PMID: 23553419 DOI: 10.1007/s00424-013-1256-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Establishment of the immunological synapse (IS) between T lymphocytes and antigen-presenting cells is a key step in the adaptive immune response. Several proteins accumulate in the IS, such as the Kv1.3 potassium channel; however, the mechanism of this translocation is unknown. PSD-95 and SAP97 are adaptor proteins that regulate the polarized cell surface expression and localization of Kv1 channels in neurons. We investigated whether these proteins affect the redistribution of Kv1.3 into the IS in non-excitable human T cells. We show here that PSD-95 and SAP97 are expressed in Jurkat and interact with the C terminus of Kv1.3. Disruption of the interaction between PSD-95 or SAP97 and Kv1.3 in Jurkat was realized by the expression of a C-terminal truncated Kv1.3, which lacks the binding domain for these proteins, or by the knockdown of the expression of PSD-95 or SAP97 using specific shRNA. Expression of the truncated Kv1.3 or knockdown of PSD-95, but not the knockdown of SAP97, inhibited the recruitment of Kv1.3 into the IS; the fraction of cells showing polarized Kv1.3 expression upon engagement in an IS was significantly lower than in control cells expressing the full-length Kv1.3, and the rearrangement of Kv1.3 did not show time dependence. In contrast, Jurkat cells expressing the full-length channel showed marked time dependence in the recruitment into the IS peaking at 1 min after the conjugation of the cells. These results demonstrate that PSD-95 participates in the targeting of Kv1.3 into the IS, implying its important role in human T-cell activation.
Collapse
|
20
|
Akama KT, Thompson LI, Milner TA, McEwen BS. Post-synaptic density-95 (PSD-95) binding capacity of G-protein-coupled receptor 30 (GPR30), an estrogen receptor that can be identified in hippocampal dendritic spines. J Biol Chem 2013; 288:6438-50. [PMID: 23300088 DOI: 10.1074/jbc.m112.412478] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The estrogen 17β-estradiol (E2) modulates dendritic spine plasticity in the cornu ammonis 1 (CA1) region of the hippocampus, and GPR30 (G-protein coupled estrogen receptor 1 (GPER1)) is an estrogen-sensitive G-protein-coupled receptor (GPCR) that is expressed in the mammalian brain and in specific subregions that are responsive to E2, including the hippocampus. The subcellular localization of hippocampal GPR30, however, remains unclear. Here, we demonstrate that GPR30 immunoreactivity is detected in dendritic spines of rat CA1 hippocampal neurons in vivo and that GPR30 protein can be found in rat brain synaptosomes. GPR30 immunoreactivity is identified at the post-synaptic density (PSD) and in the adjacent peri-synaptic zone, and GPR30 can associate with the spine scaffolding protein PSD-95 both in vitro and in vivo. This PSD-95 binding capacity of GPR30 is specific and determined by the receptor C-terminal tail that is both necessary and sufficient for PSD-95 interaction. The interaction with PSD-95 functions to increase GPR30 protein levels residing at the plasma membrane surface. GPR30 associates with the N-terminal tandem pair of PDZ domains in PSD-95, suggesting that PSD-95 may be involved in clustering GPR30 with other receptors in the hippocampus. We demonstrate that GPR30 has the potential to associate with additional post-synaptic GPCRs, including the membrane progestin receptor, the corticotropin releasing hormone receptor, and the 5HT1a serotonin receptor. These data demonstrate that GPR30 is well positioned in the dendritic spine compartment to integrate E2 sensitivity directly onto multiple inputs on synaptic activity and might begin to provide a molecular explanation as to how E2 modulates dendritic spine plasticity.
Collapse
Affiliation(s)
- Keith T Akama
- Laboratory of Neuroendocrinology, The Rockefeller University, Weill Cornell Medical College, New York, New York 10065, USA
| | | | | | | |
Collapse
|
21
|
Balse E, Steele DF, Abriel H, Coulombe A, Fedida D, Hatem SN. Dynamic of Ion Channel Expression at the Plasma Membrane of Cardiomyocytes. Physiol Rev 2012; 92:1317-58. [DOI: 10.1152/physrev.00041.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac myocytes are characterized by distinct structural and functional entities involved in the generation and transmission of the action potential and the excitation-contraction coupling process. Key to their function is the specific organization of ion channels and transporters to and within distinct membrane domains, which supports the anisotropic propagation of the depolarization wave. This review addresses the current knowledge on the molecular actors regulating the distinct trafficking and targeting mechanisms of ion channels in the highly polarized cardiac myocyte. In addition to ubiquitous mechanisms shared by other excitable cells, cardiac myocytes show unique specialization, illustrated by the molecular organization of myocyte-myocyte contacts, e.g., the intercalated disc and the gap junction. Many factors contribute to the specialization of the cardiac sarcolemma and the functional expression of cardiac ion channels, including various anchoring proteins, motors, small GTPases, membrane lipids, and cholesterol. The discovery of genetic defects in some of these actors, leading to complex cardiac disorders, emphasizes the importance of trafficking and targeting of ion channels to cardiac function. A major challenge in the field is to understand how these and other actors work together in intact myocytes to fine-tune ion channel expression and control cardiac excitability.
Collapse
Affiliation(s)
- Elise Balse
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David F. Steele
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Alain Coulombe
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David Fedida
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Stéphane N. Hatem
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Kuras Z, Kucher V, Gordon SM, Neumeier L, Chimote AA, Filipovich AH, Conforti L. Modulation of Kv1.3 channels by protein kinase A I in T lymphocytes is mediated by the disc large 1-tyrosine kinase Lck complex. Am J Physiol Cell Physiol 2012; 302:C1504-12. [PMID: 22378744 DOI: 10.1152/ajpcell.00263.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The cAMP/PKA signaling system constitutes an inhibitory pathway in T cells and, although its biochemistry has been thoroughly investigated, its possible effects on ion channels are still not fully understood. K(V)1.3 channels play an important role in T-cell activation, and their inhibition suppresses T-cell function. It has been reported that PKA modulates K(V)1.3 activity. Two PKA isoforms are expressed in human T cells: PKAI and PKAII. PKAI has been shown to inhibit T-cell activation via suppression of the tyrosine kinase Lck. The aim of this study was to determine the PKA isoform modulating K(V)1.3 and the signaling pathway underneath. 8-Bromoadenosine 3',5'-cyclic monophosphate (8-BrcAMP), a nonselective activator of PKA, inhibited K(V)1.3 currents both in primary human T and in Jurkat cells. This inhibition was prevented by the PKA blocker PKI(6-22). Selective knockdown of PKAI, but not PKAII, with siRNAs abolished the response to 8-BrcAMP. Additional studies were performed to determine the signaling pathway mediating PKAI effect on K(V)1.3. Overexpression of a constitutively active mutant of Lck reduced the response of K(V)1.3 to 8-Br-cAMP. Moreover, knockdown of the scaffolding protein disc large 1 (Dlg1), which binds K(V)1.3 to Lck, abolished PKA modulation of K(V)1.3 channels. Immunohistochemistry studies showed that PKAI, but not PKAII, colocalizes with K(V)1.3 and Dlg1 indicating a close proximity between these proteins. These results indicate that PKAI selectively regulates K(V)1.3 channels in human T lymphocytes. This effect is mediated by Lck and Dlg1. We thus propose that the K(V)1.3/Dlg1/Lck complex is part of the membrane pathway that cAMP utilizes to regulate T-cell function.
Collapse
Affiliation(s)
- Zerrin Kuras
- Department of Internal Medicine, 231 Albert Sabin Way, University of Cincinnati, Cincinnati, OH 45267-0585, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Self-directed assembly and clustering of the cytoplasmic domains of inwardly rectifying Kir2.1 potassium channels on association with PSD-95. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:2374-89. [DOI: 10.1016/j.bbamem.2011.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 06/22/2011] [Accepted: 06/28/2011] [Indexed: 12/21/2022]
|
24
|
Joseph BK, Thakali KM, Pathan AR, Kang E, Rusch NJ, Rhee SW. Postsynaptic density-95 scaffolding of Shaker-type K⁺ channels in smooth muscle cells regulates the diameter of cerebral arteries. J Physiol 2011; 589:5143-52. [PMID: 21911612 DOI: 10.1113/jphysiol.2011.213843] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Postsynaptic density-95 (PSD95) is a 95 kDa scaffolding molecule in the brain that clusters postsynaptic proteins including ion channels, receptors, enzymes and other signalling partners required for normal cognition. The voltage-gated, Shaker-type K(+) (K(V)1) channel is one key binding partner of PSD95 scaffolds in neurons. However, K(V)1 channels composed of α1.2 and α1.5 pore-forming subunits also are expressed in the vascular smooth muscle cells (cVSMCs) of the cerebral circulation, although the identity of their molecular scaffolds is unknown. Since α1.2 contains a binding motif for PSD95, we explored the possibility that cVSMCs express PSD95 as a scaffold to promote K(V)1 channel expression and cerebral vasodilatation. Cerebral arteries from Sprague-Dawley rats were isolated for analysis of PSD95 and K(V)1 channel proteins. PSD95 was detected in cVSMCs and it co-immunoprecipitated and co-localized with the pore-forming α1.2 subunit of the K(V)1 channel. Antisense-mediated knockdown of PSD95 profoundly reduced K(V)1 channel expression and suppressed K(V)1 current in patch-clamped cVSMCs. Loss of PSD95 also depolarized cVSMCs in pressurized cerebral arteries and induced a strong constriction associated with a loss of functional K(V)1 channels. Our findings provide initial evidence that PSD95 is expressed in cVSMCs, and the K(V)1 channel is one of its important binding partners. PSD95 appears to function as a critical 'dilator' scaffold in cerebral arteries by increasing the number of functional K(V)1 channels at the plasma membrane.
Collapse
Affiliation(s)
- Biny K Joseph
- Department of Pharmacology and Toxicology, College of Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ting YK, Morikawa K, Kurata Y, Li P, Bahrudin U, Mizuta E, Kato M, Miake J, Yamamoto Y, Yoshida A, Murata M, Inoue T, Nakai A, Shiota G, Higaki K, Nanba E, Ninomiya H, Shirayoshi Y, Hisatome I. Transcriptional activation of the anchoring protein SAP97 by heat shock factor (HSF)-1 stabilizes K(v) 1.5 channels in HL-1 cells. Br J Pharmacol 2011; 162:1832-42. [PMID: 21232033 PMCID: PMC3081125 DOI: 10.1111/j.1476-5381.2011.01204.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND AND PURPOSE The expression of voltage-dependent K+ channels (Kv) 1.5 is regulated by members of the heat shock protein (Hsp) family. We examined whether the heat shock transcription factor 1 (HSF-1) and its inducer geranylgeranylacetone (GGA) could affect the expression of Kv1.5 channels and its anchoring protein, synapse associated protein 97 (SAP97). EXPERIMENTAL APPROACH Transfected mouse atrial cardiomyocytes (HL-1 cells) and COS7 cells were subjected to luciferase reporter gene assay and whole-cell patch clamp. Protein and mRNA extracts were subjected to Western blot and quantitative real-time polymerase chain reaction. KEY RESULTS Heat shock of HL-1 cells induced expression of Hsp70, HSF-1, SAP97 and Kv1.5 proteins. These effects were reproduced by wild-type HSF-1. Both heat shock and expression of HSF-1, but not the R71G mutant, increased the SAP97 mRNA level. Small interfering RNA (siRNA) against SAP97 abolished HSF-1-induced increase of Kv1.5 and SAP97 proteins. A luciferase reporter gene assay revealed that the SAP97 promoter region (from −919 to −740) that contains heat shock elements (HSEs) was required for this induction. Suppression of SIRT1 function either by nicotinamide or siRNA decreased the level of SAP97 mRNA. SIRT1 activation by resveratrol had opposing effects. A treatment of the cells with GGA increased the level of SAP97 mRNA, Kv1.5 proteins and IKur current, which could be modified with either resveratrol or nicotinamide. CONCLUSIONS AND IMPLICATIONS HSF-1 induced transcription of SAP97 through SIRT1-dependent interaction with HSEs; the increase in SAP97 resulted in stabilization of Kv1.5 channels. These effects were mimicked by GGA.
Collapse
Affiliation(s)
- Y K Ting
- Division of Regenerative Medicine and Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science, Yonago, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
D'Amico A, Soragna A, Di Cairano E, Panzeri N, Anzai N, Vellea Sacchi F, Perego C. The surface density of the glutamate transporter EAAC1 is controlled by interactions with PDZK1 and AP2 adaptor complexes. Traffic 2010; 11:1455-70. [PMID: 20727120 DOI: 10.1111/j.1600-0854.2010.01110.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The glutamate transporter excitatory amino acid carrier (EAAC1/EAAT3) mediates the absorption of dicarboxylic amino acids in epithelial cells as well as the uptake of glutamate from the synaptic cleft. Its cell-surface density is regulated by interaction with accessory proteins which remain to be identified. We detected a consensus sequence for interaction with post-synaptic density-95/Discs large/Zonula occludens (PDZ) proteins (-SQF) and a tyrosine-based internalization signal (-YVNG-) in the C-terminus of EAAC1, and investigated their role in the transporter localization. We demonstrated that PDZ interactions are required for the efficient delivery to and the retention in the plasma membrane of EAAC1 and we identified PDZK1/NHERF3 (Na+/H+-exchanger regulatory factor 3) as a novel EAAC1 interacting protein. Expression of PDZK1 in Madin-Darby canine kidney (MDCK) cells tethered EAAC1 to filopodia and increased its surface activity. Removal of the PDZ-target motif promoted the EAAC1 binding to α-adaptin and clathrin and the transporter internalization in endocytic/degradative compartments. This defect was largely prevented by hypertonic treatment or overexpression of the dominant-negative µ2-W421A-subunit of AP-2 clathrin-adaptor. The rate of transporter endocytosis was attenuated following tyrosine mutagenesis in the internalization signal, thus indicating that this motif can regulate the transporter endocytosis. We suggest that EAAC1 density is controlled by balanced interactions with PDZK1 and adaptor protein 2 (AP2): the former promotes the transporter expression at the cell surface, and the latter mediates its constitutive endocytosis.
Collapse
Affiliation(s)
- Anna D'Amico
- Department of Molecular Sciences Applied to Biosystems, Laboratory of Cellular Physiology, Università degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Vaidyanathan R, Taffet SM, Vikstrom KL, Anumonwo JMB. Regulation of cardiac inward rectifier potassium current (I(K1)) by synapse-associated protein-97. J Biol Chem 2010; 285:28000-9. [PMID: 20530486 DOI: 10.1074/jbc.m110.110858] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Synapse-associated protein-97 (SAP97) is a membrane-associated guanylate kinase scaffolding protein expressed in cardiomyocytes. SAP97 has been shown to associate and modulate voltage-gated potassium (Kv) channel function. In contrast to Kv channels, little information is available on interactions involving SAP97 and inward rectifier potassium (Kir2.x) channels that underlie the classical inward rectifier current, I(K1). To investigate the functional effects of silencing SAP97 on I(K1) in adult rat ventricular myocytes, SAP97 was silenced using an adenoviral short hairpin RNA vector. Western blot analysis showed that SAP97 was silenced by approximately 85% on day 3 post-infection. Immunostaining showed that Kir2.1 and Kir2.2 co-localize with SAP97. Co-immunoprecipitation (co-IP) results demonstrated that Kir2.x channels associate with SAP97. Voltage clamp experiments showed that silencing SAP97 reduced I(K1) whole cell density by approximately 55%. I(K1) density at -100 mV was -1.45 +/- 0.15 pA/picofarads (n = 6) in SAP97-silenced cells as compared with -3.03 +/- 0.37 pA/picofarads (n = 5) in control cells. Unitary conductance properties of I(K1) were unaffected by SAP97 silencing. The major mechanism for the reduction of I(K1) density appears to be a decrease in Kir2.x channel abundance. Furthermore, SAP97 silencing impaired I(K1) regulation by beta(1)-adrenergic receptor (beta1-AR) stimulation. In control, isoproterenol reduced I(K1) amplitude by approximately 75%, an effect that was blunted following SAP97 silencing. Our co-IP data show that beta1-AR associates with SAP97 and Kir2.1 and also that Kir2.1 co-IPs with protein kinase A and beta1-AR. SAP97 immunolocalizes with protein kinase A and beta1-AR in the cardiac myocytes. Our results suggest that in cardiac myocytes SAP97 regulates surface expression of channels underlying I(K1), as well as assembles a signaling complex involved in beta1-AR regulation of I(K1).
Collapse
Affiliation(s)
- Ravi Vaidyanathan
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York 13210, USA
| | | | | | | |
Collapse
|
28
|
Raja M, Vales E. Mutations in the K(+)-channel KcsA toward Kir channels alter salt-induced clusterization and blockade by quaternary alkylammonium ions. J Membr Biol 2010; 233:43-50. [PMID: 20037756 DOI: 10.1007/s00232-009-9223-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 12/02/2009] [Indexed: 11/26/2022]
Abstract
Protein aggregation is a result of malfunction in protein folding, assembly, and transport, caused by protein mutation and/or changes in the cell environment, thus triggering many human diseases. We have shown that bacterial K(+)-channel KcsA, which acts as a representative model for ion channels, forms salt-induced large conductive complexes in a particular environment. In the present study, we investigated the effects of point mutations in the selectivity filter of KcsA on intrinsic stability, aggregation, and channel blocking behavior. First, we found that a low sodium chloride concentration in potassium-containing media induced fast transfer of single channels to a planar lipid bilayer. Second, increasing the sodium chloride concentration drastically increased the total channel current, indicating enhanced vesicle fusion and transfer of multiple channels to a planar lipid bilayer. However, such complexes exhibited high conductance as well as higher open probability compared to the unmodified KcsA behavior shown previously. Interestingly, the affinity of aggregated complexes for larger symmetric quaternary alkylammonium ions (QAs) was found to be much higher than that for tetraethylammonium, a classical blocker of the K(+) channel. Based on these findings, we propose that mutant channel complexes exhibit larger pore dimensions, thus resembling more the topological properties of voltage-gated and inwardly rectifying K(+) channels.
Collapse
Affiliation(s)
- Mobeen Raja
- Institute for Biophysics, Johannes Kepler University Linz, Austria.
| | | |
Collapse
|
29
|
Nicolaou SA, Neumeier L, Steckly A, Kucher V, Takimoto K, Conforti L. Localization of Kv1.3 channels in the immunological synapse modulates the calcium response to antigen stimulation in T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2009; 183:6296-302. [PMID: 19841189 DOI: 10.4049/jimmunol.0900613] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The immunological synapse (IS), a highly organized structure that forms at the point of contact between a T cell and an APC, is essential for the proper development of signaling events, including the Ca(2+) response. Kv1.3 channels control Ca(2+) homeostasis in human T cells and move into the IS upon Ag presentation. However, the process involved in channel accumulation in the IS and the functional implications of this localization are not yet known. Here we define the movement of Kv1.3 into the IS and study whether Kv1.3 localization into the IS influences Ca(2+) signaling in Jurkat T cells. Crosslinking of the channel protein with an extracellular Ab limits Kv1.3 mobility and accumulation at the IS. Moreover, Kv1.3 recruitment to the IS does not involve the transport of newly synthesized channels and it does not occur through recycling of membrane channels. Kv1.3 localization in the IS modulates the Ca(2+) response. Blockade of Kv1.3 movement into the IS by crosslinking significantly increases the amplitude of the Ca(2+) response triggered by anti-CD3/anti-CD28-coated beads, which induce the formation of the IS. On the contrary, the Ca(2+) response induced by TCR stimulation without the formation of the IS with soluble anti-CD3/anti-CD28 Abs is unaltered. The results presented herein indicate that, upon Ag presentation, membrane-incorporated Kv1.3 channels move along the plasma membrane to localize in the IS. This localization is important to control the amplitude of the Ca(2+) response, and disruption of this process can account for alterations of downstream Ca(2+)-dependent signaling events.
Collapse
Affiliation(s)
- Stella A Nicolaou
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | | | | | | | |
Collapse
|
30
|
Kruse M, Schulze-Bahr E, Corfield V, Beckmann A, Stallmeyer B, Kurtbay G, Ohmert I, Schulze-Bahr E, Brink P, Pongs O. Impaired endocytosis of the ion channel TRPM4 is associated with human progressive familial heart block type I. J Clin Invest 2009; 119:2737-44. [PMID: 19726882 DOI: 10.1172/jci38292] [Citation(s) in RCA: 236] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Accepted: 06/10/2009] [Indexed: 01/20/2023] Open
Abstract
Progressive familial heart block type I (PFHBI) is a progressive cardiac bundle branch disease in the His-Purkinje system that exhibits autosomal-dominant inheritance. In 3 branches of a large South African Afrikaner pedigree with an autosomal-dominant form of PFHBI, we identified the mutation c.19G-->A in the transient receptor potential cation channel, subfamily M, member 4 gene (TRPM4) at chromosomal locus 19q13.3. This mutation predicted the amino acid substitution p.E7K in the TRPM4 amino terminus. TRPM4 encodes a Ca2+-activated nonselective cation (CAN) channel that belongs to the transient receptor potential melastatin ion channel family. Quantitative analysis of TRPM4 mRNA content in human cardiac tissue showed the highest expression level in Purkinje fibers. Cellular expression studies showed that the c.19G-->A missense mutation attenuated deSUMOylation of the TRPM4 channel. The resulting constitutive SUMOylation of the mutant TRPM4 channel impaired endocytosis and led to elevated TRPM4 channel density at the cell surface. Our data therefore revealed a gain-of-function mechanism underlying this type of familial heart block.
Collapse
Affiliation(s)
- Martin Kruse
- Institut für Neurale Signalverarbeitung, Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Vikstrom KL, Vaidyanathan R, Levinsohn S, O'Connell RP, Qian Y, Crye M, Mills JH, Anumonwo JMB. SAP97 regulates Kir2.3 channels by multiple mechanisms. Am J Physiol Heart Circ Physiol 2009; 297:H1387-97. [PMID: 19633205 DOI: 10.1152/ajpheart.00638.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We examined the impact of coexpressing the inwardly rectifying potassium channel, Kir2.3, with the scaffolding protein, synapse-associated protein (SAP) 97, and determined that coexpression of these proteins caused an approximately twofold increase in current density. A combination of techniques was used to determine if the SAP97-induced increase in Kir2.3 whole cell currents resulted from changes in the number of channels in the cell membrane, unitary channel conductance, or channel open probability. In the absence of SAP97, Kir2.3 was found predominantly in a cytoplasmic, vesicular compartment with relatively little Kir2.3 localized to the plasma membrane. The introduction of SAP97 caused a redistribution of Kir2.3, leading to prominent colocalization of Kir2.3 and SAP97 and a modest increase in cell surface Kir2.3. The median Kir2.3 single channel conductance in the absence of SAP97 was approximately 13 pS, whereas coexpression of SAP97 led to a wide distribution of channel events with three distinct peaks centered at 16, 29, and 42 pS. These changes occurred without altering channel open probability, current rectification properties, or pH sensitivity. Thus association of Kir2.3 with SAP97 in HEK293 cells increased channel cell surface expression and unitary channel conductance. However, changes in single channel conductance play the major role in determining whole cell currents in this model system. We further suggest that the SAP97 effect results from SAP97 binding to the Kir2.3 COOH-terminal domain and altering channel conformation.
Collapse
Affiliation(s)
- Karen L Vikstrom
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Niwa N, Nerbonne JM. Molecular determinants of cardiac transient outward potassium current (I(to)) expression and regulation. J Mol Cell Cardiol 2009; 48:12-25. [PMID: 19619557 DOI: 10.1016/j.yjmcc.2009.07.013] [Citation(s) in RCA: 167] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/25/2009] [Accepted: 07/10/2009] [Indexed: 12/21/2022]
Abstract
Rapidly activating and inactivating cardiac transient outward K(+) currents, I(to), are expressed in most mammalian cardiomyocytes, and contribute importantly to the early phase of action potential repolarization and to plateau potentials. The rapidly recovering (I(t)(o,f)) and slowly recovering (I(t)(o,s)) components are differentially expressed in the myocardium, contributing to regional heterogeneities in action potential waveforms. Consistent with the marked differences in biophysical properties, distinct pore-forming (alpha) subunits underlie the two I(t)(o) components: Kv4.3/Kv4.2 subunits encode I(t)(o,f), whereas Kv1.4 encodes I(t)(o,s), channels. It has also become increasingly clear that cardiac I(t)(o) channels function as components of macromolecular protein complexes, comprising (four) Kvalpha subunits and a variety of accessory subunits and regulatory proteins that influence channel expression, biophysical properties and interactions with the actin cytoskeleton, and contribute to the generation of normal cardiac rhythms. Derangements in the expression or the regulation of I(t)(o) channels in inherited or acquired cardiac diseases would be expected to increase the risk of potentially life-threatening cardiac arrhythmias. Indeed, a recently identified Brugada syndrome mutation in KCNE3 (MiRP2) has been suggested to result in increased I(t)(o,f) densities. Continued focus in this area seems certain to provide new and fundamentally important insights into the molecular determinants of functional I(t)(o) channels and into the molecular mechanisms involved in the dynamic regulation of I(t)(o) channel functioning in the normal and diseased myocardium.
Collapse
Affiliation(s)
- Noriko Niwa
- Department of Developmental Biology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8103, St. Louis, MO 63110-1093, USA
| | | |
Collapse
|
33
|
PSD-95 mediates membrane clustering of the human plasma membrane Ca2+ pump isoform 4b. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:1023-32. [PMID: 19073225 DOI: 10.1016/j.bbamcr.2008.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Revised: 10/31/2008] [Accepted: 11/16/2008] [Indexed: 01/09/2023]
Abstract
Besides the control of global calcium changes, specific plasma membrane calcium ATPase (PMCA) isoforms are involved in the regulation of local calcium signals. Although local calcium signaling requires the confinement of signaling molecules into microdomains, little is known about the specific organization of PMCA molecules within the plasma membrane. Here we show that co-expression with the postsynaptic density-95 (PSD-95) scaffolding protein increased the plasma membrane expression of PMCA4b and redistributed the pump into clusters. The clustering of PMCA4b was fully dependent on the presence of its PDZ-binding sequence. Using the fluorescence recovery after photobleaching (FRAP) technique, we show that the lateral membrane mobility of the clustered PMCA4b is significantly lower than that of the non-clustered molecules. Disruption of the actin-based cytoskeleton by cytochalasin D resulted in increased cluster size. Our results suggest that PSD-95 promotes the formation of high-density PMCA4b microdomains in the plasma membrane and that the membrane cytoskeleton plays an important role in the regulation of this process.
Collapse
|
34
|
Mckeown L, Burnham MP, Hodson C, Jones OT. Identification of an evolutionarily conserved extracellular threonine residue critical for surface expression and its potential coupling of adjacent voltage-sensing and gating domains in voltage-gated potassium channels. J Biol Chem 2008; 283:30421-32. [PMID: 18640987 DOI: 10.1074/jbc.m708921200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The dynamic expression of voltage-gated potassium channels (Kvs) at the cell surface is a fundamental factor controlling membrane excitability. In exploring possible mechanisms controlling Kv surface expression, we identified a region in the extracellular linker between the first and second of the six (S1-S6) transmembrane-spanning domains of the Kv1.4 channel, which we hypothesized to be critical for its biogenesis. Using immunofluorescence microscopy, flow cytometry, patch clamp electrophysiology, and mutagenesis, we identified a single threonine residue at position 330 within the Kv1.4 S1-S2 linker that is absolutely required for cell surface expression. Mutation of Thr-330 to an alanine, aspartate, or lysine prevented surface expression. However, surface expression occurred upon co-expression of mutant and wild type Kv1.4 subunits or mutation of Thr-330 to a serine. Mutation of the corresponding residue (Thr-211) in Kv3.1 to alanine also caused intracellular retention, suggesting that the conserved threonine plays a generalized role in surface expression. In support of this idea, sequence comparisons showed conservation of the critical threonine in all Kv families and in organisms across the evolutionary spectrum. Based upon the Kv1.2 crystal structure, further mutagenesis, and the partial restoration of surface expression in an electrostatic T330K bridging mutant, we suggest that Thr-330 hydrogen bonds to equally conserved outer pore residues, which may include a glutamate at position 502 that is also critical for surface expression. We propose that Thr-330 serves to interlock the voltage-sensing and gating domains of adjacent monomers, thereby yielding a structure competent for the surface expression of functional tetramers.
Collapse
Affiliation(s)
- Lynn Mckeown
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PN, United Kingdom
| | | | | | | |
Collapse
|
35
|
McKeown L, Swanton L, Robinson P, Jones OT. Surface expression and distribution of voltage-gated potassium channels in neurons (Review). Mol Membr Biol 2008; 25:332-43. [PMID: 18446619 DOI: 10.1080/09687680801992470] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed an exponential increase in interest in one of the great mysteries of nerve cell biology: Specifically, how do neurons know where to place the ion channels that control their excitability? Many of the most important insights have been gleaned from studies on the voltage-gated potassium channels (Kvs) which underlie the shape, duration and frequency of action potentials. In this review, we gather recent evidence on the expression, trafficking and maintenance mechanisms which control the surface density of Kvs in different subcellular compartments of neurons and how these may be regulated to control cell excitability.
Collapse
Affiliation(s)
- Lynn McKeown
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
36
|
González-González IM, García-Tardón N, Cubelos B, Giménez C, Zafra F. The glutamate transporter GLT1b interacts with the scaffold protein PSD-95. J Neurochem 2008; 105:1834-48. [DOI: 10.1111/j.1471-4159.2008.05281.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
37
|
Liu G, Zakharov SI, Yang L, Deng SX, Landry DW, Karlin A, Marx SO. Position and role of the BK channel alpha subunit S0 helix inferred from disulfide crosslinking. ACTA ACUST UNITED AC 2008; 131:537-48. [PMID: 18474637 PMCID: PMC2391248 DOI: 10.1085/jgp.200809968] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The position and role of the unique N-terminal transmembrane (TM) helix, S0, in large-conductance, voltage- and calcium-activated potassium (BK) channels are undetermined. From the extents of intra-subunit, endogenous disulfide bond formation between cysteines substituted for the residues just outside the membrane domain, we infer that the extracellular flank of S0 is surrounded on three sides by the extracellular flanks of TM helices S1 and S2 and the four-residue extracellular loop between S3 and S4. Eight different double cysteine-substituted alphas, each with one cysteine in the S0 flank and one in the S3-S4 loop, were at least 90% disulfide cross-linked. Two of these alphas formed channels in which 90% cross-linking had no effect on the V(50) or on the activation and deactivation rate constants. This implies that the extracellular ends of S0, S3, and S4 are close in the resting state and move in concert during voltage sensor activation. The association of S0 with the gating charge bearing S3 and S4 could contribute to the considerably larger electrostatic energy required to activate the BK channel compared with typical voltage-gated potassium channels with six TM helices.
Collapse
Affiliation(s)
- Guoxia Liu
- 1 Division of Cardiology, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Abi-Char J, El-Haou S, Balse E, Neyroud N, Vranckx R, Coulombe A, Hatem SN. The anchoring protein SAP97 retains Kv1.5 channels in the plasma membrane of cardiac myocytes. Am J Physiol Heart Circ Physiol 2008; 294:H1851-61. [DOI: 10.1152/ajpheart.01045.2007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Membrane- associated guanylate kinase proteins (MAGUKs) are important determinants of localization and organization of ion channels into specific plasma membrane domains. However, their exact role in channel function and cardiac excitability is not known. We examined the effect of synapse-associated protein 97 (SAP97), a MAGUK abundantly expressed in the heart, on the function and localization of Kv1.5 subunits in cardiac myocytes. Recombinant SAP97 or Kv1.5 subunits tagged with green fluorescent protein (GFP) were overexpressed in rat neonatal cardiac myocytes and in Chinese hamster ovary (CHO) cells from adenoviral or plasmidic vectors. Immunocytochemistry, fluorescence recovery after photobleaching, and patch-clamp techniques were used to study the effects of SAP97 on the localization, mobility, and function of Kv1.5 subunits. Adenovirus-mediated SAP97 overexpression in cardiac myocytes resulted in the clustering of endogenous Kv1.5 subunits at myocyte-myocyte contacts and an increase in both the maintained component of the outward K+current, IKur(5.64 ± 0.57 pA/pF in SAP97 myocytes vs. 3.23 ± 0.43 pA/pF in controls) and the number of 4-aminopyridine-sensitive potassium channels in cell-attached membrane patches. In live myocytes, GFP-Kv1.5 subunits were mobile and organized in clusters at the basal plasma membrane, whereas SAP97 overexpression reduced their mobility. In CHO cells, Kv1.5 channels were diffusely distributed throughout the cell body and freely mobile. When coexpressed with SAP97, Kv subunits were organized in plaquelike clusters and poorly mobile. In conclusion, SAP97 regulates the K+current in cardiac myocytes by retaining and immobilizing Kv1.5 subunits in the plasma membrane. This new regulatory mechanism may contribute to the targeting of Kv channels in cardiac myocytes.
Collapse
|
39
|
Jindal HK, Folco EJ, Liu GX, Koren G. Posttranslational modification of voltage-dependent potassium channel Kv1.5: COOH-terminal palmitoylation modulates its biological properties. Am J Physiol Heart Circ Physiol 2008; 294:H2012-21. [PMID: 18344374 DOI: 10.1152/ajpheart.01374.2007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The physiological function of ion channels is affected by protein-protein and protein-membrane interactions that modulate their activity and/or localization. Palmitoylation modulates protein function by facilitating targeted membrane association, interaction with other proteins, and determining subcellular localization. In this study, we demonstrate that the voltage-dependent potassium (Kv) channel Kv1.5 is palmitoylated and that the mutation of COOH-terminal cysteines is sufficient to abolish the palmitoylation of the Kv1.5 polypeptide in Chinese hamster ovary (CHO) cells. The labeling represented the thioester linkage of the labeled palmitic acid to cysteine rather than amide and oxygen ester linkages as judged by the release of the palmitic acid upon the treatment of the gel with hydroxylamine at a neutral pH. Site-directed mutagenesis and radiolabeling studies revealed that C593 was the sole site of palmitoylation. The elucidation of the biological function of palmitoylation revealed that the expression of the FLAG-Kv1.5 palmitoylation-deficient mutant (FL-Kv1.5(Palm-)) in stable CHO cells increased membrane expression as determined by the biotinylation of surface proteins and quantitative immunofluorescence analyses of these cells, in turn enhancing the outward potassium current. This enhanced surface expression and the currents were consequential to the slower rate of internalization, causing an increased localization of FL-Kv1.5(Palm-) in the plasma membrane compared with the wild-type FL-Kv1.5 channels. We conclude that the Kv1.5 channel is palmitoylated and that its palmitoylation modulates its biological functions and, therefore, might provide a physiological link between the metabolic state and the expression of Kv1.5 on the plasma membrane.
Collapse
Affiliation(s)
- Hitesh K Jindal
- Cardiovascular Research Center, Rhode Island Hospital, Brown University School of Medicine, 1 Hoppin Street, Providence, RI 02903, USA
| | | | | | | |
Collapse
|
40
|
Jones VC, McKeown L, Verkhratsky A, Jones OT. LV-pIN-KDEL: a novel lentiviral vector demonstrates the morphology, dynamics and continuity of the endoplasmic reticulum in live neurones. BMC Neurosci 2008; 9:10. [PMID: 18215281 PMCID: PMC2248189 DOI: 10.1186/1471-2202-9-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Accepted: 01/23/2008] [Indexed: 11/24/2022] Open
Abstract
Background The neuronal endoplasmic reticulum (ER) is an extensive, complex endomembrane system, containing Ca2+ pumps, and Ca2+ channels that permit it to act as a dynamic calcium store. Currently, there is controversy over the continuity of the ER in neurones, how this intersects with calcium signalling and the possibility of physical compartmentalisation. Unfortunately, available probes of ER structure such as vital dyes are limited by their membrane specificity. The introduction of ER-targeted GFP plasmids has been a considerable step forward, but these are difficult to express in neurones through conventional transfection approaches. To circumvent such problems we have engineered a novel ER-targeted GFP construct, termed pIN-KDEL, into a 3rd generation replication-defective, self-inactivating lentiviral vector system capable of mediating gene transduction in diverse dividing and post-mitotic mammalian cells, including neurones. Results Following its expression in HEK293 (or COS-7) cells, LV-pIN-KDEL yielded a pattern of fluorescence that co-localised exclusively with the ER marker sec61β but with no other major organelle. We found no evidence for cytotoxicity and only rarely inclusion body formation. To explore the utility of the probe in resolving the ER in live cells, HEK293 or COS-7 cells were transduced with LV-pIN-KDEL and, after 48 h, imaged directly at intervals from 1 min to several hours. LV-pIN-KDEL fluorescence revealed the endoplasmic reticulum as a tubular lattice structure whose morphology can change markedly within seconds. Although GFP can be phototoxic, the integrity of the cells and ER was retained for several weeks and even after light exposure for periods up to 24 h. Using LV-pIN-KDEL we have imaged the ER in diverse fixed neuronal cultures and, using real-time imaging, found evidence for extensive, dynamic remodelling of the neuronal ER in live hippocampal cultures, brain slices, explants and glia. Finally, through a Fluorescence Loss in Photobleaching (FLIP) approach, continuous irradiation at a single region of interest removed all the fluorescence of LV-pIN-KDEL-transduced nerve cells in explant cultures, thus, providing compelling evidence that in neurons the endoplasmic reticulum is not only dynamic but also continuous. Conclusion The lentiviral-based ER-targeted reporter, LV-pIN-KDEL, offers considerable advantages over present systems for defining the architecture of the ER, especially in primary cells such as neurones that are notoriously difficult to transfect. Images and continuous photobleaching experiments of LV-pIN-KDEL-transduced neurones demonstrate that the endoplasmic reticulum is a dynamic structure with a single continuous lumen. The introduction of LV-pIN-KDEL is anticipated to greatly facilitate a real-time visualisation of the structural plasticity and continuous nature of the neuronal ER in healthy and diseased brain tissue.
Collapse
Affiliation(s)
- Vicky C Jones
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9NT, UK.
| | | | | | | |
Collapse
|
41
|
Marks DR, Fadool DA. Post-synaptic density perturbs insulin-induced Kv1.3 channel modulation via a clustering mechanism involving the SH3 domain. J Neurochem 2007; 103:1608-27. [PMID: 17854350 PMCID: PMC2667938 DOI: 10.1111/j.1471-4159.2007.04870.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The olfactory bulb (OB) contains the highest concentration of the insulin receptor (IR) kinase in the central nervous system; however, its functional role and modulation in this region remains poorly understood. IR kinase contains a number of proline-rich motifs, making it an excellent candidate for modulation by SH(3) domain-containing adaptor proteins. Kv1.3, a voltage-gated Shaker potassium channel and tyrosine phosphorylation substrate of IR kinase, contains several proline-rich sequences and a canonical post-synaptic density 95 (PSD-95)/discs large/zO-1 domain (PDZ) recognition motif common to most Shaker family members. We sought to determine if a functional relationship existed between Kv1.3, IR kinase, and the SH(3)/PDZ adaptor protein PSD-95. Through patch-clamp electrophysiology, immunochemistry, and co-immunoprecipitation, we found that while Kv1.3 and PSD-95 alone interact via the canonical C-terminal PDZ recognition motif of the channel, this molecular site of interaction acts to cluster the channels but the PSD-95 SH(3)-guanylate kinase domain functionally modulates Kv1.3 activity via two proline-rich domains in its N- and C-terminal. Therefore, these data suggest that adaptor domains responsible for ion-channel clustering and functional modulation are not necessarily coupled. Moreover, IR kinase and Kv1.3 can only be co-immunoprecipitated in the presence of PSD-95 as the adapting linker. Functionally, insulin-dependent Kv1.3 phosphorylation that causes channel current suppression is blocked via interaction with the PSD-95 SH(3)-guanylate kinase domain. Because all the three proteins co-localize in multiple lamina of the OB that are known to be rich in synaptic connections, membrane excitability and synaptic transmission at critical locations in the OB have the capacity to be finely regulated.
Collapse
Affiliation(s)
- D R Marks
- Department of Biological Science, Program in Neuroscience, Biomedical Research Facility, The Florida State University, Tallahassee, Florida, USA
| | | |
Collapse
|
42
|
McEwen DP, Schumacher SM, Li Q, Benson MD, Iñiguez-Lluhí JA, Van Genderen KM, Martens JR. Rab-GTPase-dependent endocytic recycling of Kv1.5 in atrial myocytes. J Biol Chem 2007; 282:29612-20. [PMID: 17673464 DOI: 10.1074/jbc.m704402200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The number of ion channels expressed on the cell surface shapes the complex electrical response of excitable cells. Maintaining a balance between anterograde and retrograde trafficking of channel proteins is vital in regulating steady-state cell surface expression. Kv1.5 is an important voltage-gated K(+) channel in the cardiovascular system underlying the ultra-rapid rectifying potassium current (Ik(ur)), a major repolarizing current in atrial myocytes, and regulating the resting membrane potential and excitability of smooth muscle cells. Defects in the expression of Kv1.5 are associated with pathological states such as chronic atrial fibrillation and hypoxic pulmonary hypertension. There is, thus, substantial interest in understanding the mechanisms regulating cell surface channel levels. Here, we investigated the internalization and recycling of Kv1.5 in the HL-1 immortalized mouse atrial myocytes. Kinetic studies indicate that Kv1.5 is rapidly internalized to a perinuclear region where it co-localizes with the early endosomal marker, EEA1. Importantly, we identified that a population of Kv1.5, originating on the cell surface, internalized and recycled back to the plasma membrane. Notably, Kv1.5 recycling processes are driven by specific Rab-dependent endosomal compartments. Thus, co-expression of GDP-locked Rab4S22N and Rab11S25N dominant-negative mutants decreased the steady-state Kv1.5 surface levels, whereas GTPase-deficient Rab4Q67L and Rab11Q70L mutants increased steady-state Kv1.5 surface levels. These data reveal an unexpected dynamic trafficking of Kv1.5 at the myocyte plasma membrane and demonstrate a role for recycling in the maintenance of steady-state ion channel surface levels.
Collapse
Affiliation(s)
- Dyke P McEwen
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Hervé JC, Derangeon M, Bahbouhi B, Mesnil M, Sarrouilhe D. The connexin turnover, an important modulating factor of the level of cell-to-cell junctional communication: comparison with other integral membrane proteins. J Membr Biol 2007; 217:21-33. [PMID: 17673963 DOI: 10.1007/s00232-007-9054-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 06/04/2007] [Indexed: 12/25/2022]
Abstract
The constituent proteins of gap junctions, called "connexins" (Cxs) in chordates, are generally renewed several times a day, in approximately the same rate range as many other integral plasma membrane proteins and the proteins of other channels, other intercellular junctions or different membrane receptors. This permanent renewal turns on a fine-tuned balance among various processes, such as gene transcription, mRNA stability and processing, protein synthesis and oligomerization, posttranslational modifications, transport to the plasma membrane, anchoring to the cytoskeleton, connexon aggregation and docking, regulation of endocytosis and controlled degradations of the proteins. Subtle changes at one or some of these steps would represent an exquisite level of regulation that extends beyond the rapid channel opening and closure events associated with channel gating; membrane channels and receptors are constantly able to answer to physiological requirements to either up- or downregulate their activity. The Cx turnover rate thereby appears to be a key component in the regulation of any protein, particularly of gap junctional proteins. However, the physiological stimuli that control the assembly of Cxs into gap junctions and their degradation remain poorly understood.
Collapse
Affiliation(s)
- Jean-Claude Hervé
- Institut de Physiologie et Biologie Cellulaires, Faculté des Sciences Fondamentales et Appliquées, UMR CNRS 6187, Université de Poitiers, 40, avenue du R Pineau, 86022, Poitiers, France.
| | | | | | | | | |
Collapse
|
44
|
Akopian AN, Ruparel NB, Jeske NA, Hargreaves KM. Transient receptor potential TRPA1 channel desensitization in sensory neurons is agonist dependent and regulated by TRPV1-directed internalization. J Physiol 2007; 583:175-93. [PMID: 17584831 PMCID: PMC2277224 DOI: 10.1113/jphysiol.2007.133231] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The pharmacological desensitization of receptors is a fundamental mechanism for regulating the activity of neuronal systems. The TRPA1 channel plays a key role in the processing of noxious information and can undergo functional desensitization by unknown mechanisms. Here we show that TRPA1 is desensitized by homologous (mustard oil; a TRPA1 agonist) and heterologous (capsaicin; a TRPV1 agonist) agonists via Ca2+-independent and Ca2+-dependent pathways, respectively, in sensory neurons. The pharmacological desensitization of TRPA1 by capsaicin and mustard oil is not influenced by activation of protein phosphatase 2B. However, it is regulated by phosphatidylinositol-4,5-bisphosphate depletion after capsaicin, but not mustard oil, application. Using a biosensor, we establish that capsaicin, unlike mustard oil, consistently activates phospholipase C in sensory neurons. We next demonstrate that TRPA1 desensitization is regulated by TRPV1, and it appears that mustard oil-induced TRPA1 internalization is prevented by coexpression with TRPV1 in a heterologous expression system and in sensory neurons. In conclusion, we propose novel mechanisms whereby TRPA1 activity undergoes pharmacological desensitization through multiple cellular pathways that are agonist dependent and modulated by TRPV1.
Collapse
Affiliation(s)
- Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA.
| | | | | | | |
Collapse
|
45
|
Hervé JC, Bourmeyster N, Sarrouilhe D, Duffy HS. Gap junctional complexes: From partners to functions. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2007; 94:29-65. [PMID: 17507078 DOI: 10.1016/j.pbiomolbio.2007.03.010] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gap junctions (GJ), specialised membrane structures that mediate cell-to-cell communication in almost all animal tissues, are composed of intercellular channel-forming integral membrane proteins termed connexins (Cxs), innexins or pannexins. The activity of these channels is closely regulated, particularly by intramolecular modifications as phosphorylation of proteins, via the formation of multiprotein complexes where pore-forming subunits bind to auxiliary channel subunits and associate with scaffolding proteins that play essential roles in channel localization and activity. Scaffolding proteins link signalling enzymes, substrates, and potential effectors (such as channels) into multiprotein signalling complexes that may be anchored to the cytoskeleton. Protein-protein interactions play essential roles in channel localization and activity and, besides their cell-to-cell channel-forming functions, gap junctional proteins now appear involved in different cellular functions (e.g. transcriptional and cytoskeletal regulation). The present review summarizes the recent progress regarding the proteins capable of interacting with junctional proteins and their functional importance.
Collapse
Affiliation(s)
- Jean-Claude Hervé
- Interactions et Communications Cellulaires, Université de Poitiers, Poitiers, France.
| | | | | | | |
Collapse
|
46
|
Bernstein GM, Jones OT. Kinetics of internalization and degradation of N-type voltage-gated calcium channels: Role of the α2/δ subunit. Cell Calcium 2007; 41:27-40. [PMID: 16759698 DOI: 10.1016/j.ceca.2006.04.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Revised: 04/22/2006] [Accepted: 04/24/2006] [Indexed: 11/27/2022]
Abstract
The contribution of voltage-gated calcium channels to excitable cell function depends, critically, upon the mechanisms that control their expression at the cell surface. While co-assembly of the pore forming alpha(1) and auxiliary beta subunits enhances channel surface expression, the levels are still only 30-40% of those seen with the core alpha(1B)/beta(1b)/alpha(2)delta calcium channel complex. To rationalize this observation, it has been suggested that the alpha(2)/delta subunit might stabilize calcium channel expression at the cell surface. To test this notion, we have resolved the effect of the alpha(2)/delta subunit on the rates of binding, internalization and degradation of defined N-type calcium channel surface complexes expressed in HEK293 cells, through pulse-labeling with the selective, cell impermeable, radioligand [(125)I]-omega-CgTx. Through detailed kinetic and sensitivity analysis we show that alpha(1B)/beta(1b)/alpha(2)delta complexes are internalized slowly (k(int) 0.4/h), whereupon, most become degraded (k(deg) 0.02/h). In contrast, alpha(1B)/beta(1b) complexes are internalized more rapidly (k(int) 0.8/h), following which they are either quickly degraded (k(deg) 0.1/h) or are sequestered slowly (k(tra) 0.1/h) to a pool that is metabolically stable within the time-frame of our experiments (24h). In neither case did we find evidence for recycling via the cell surface. Thus, our data argue for a novel mechanism where complexes lacking an alpha(2)/delta subunit are cleared from the cell surface and are rapidly degraded or stored, possibly for further attempts at complexation as new alpha(2)/delta subunits become available. The slower rate of internalization of complexes containing the alpha(2)/delta subunit rationalizes the stabilizing effect this subunit has upon calcium channel surface expression and suggests a mechanism by which alpha(2)delta mutations may cause severe neurological deficits.
Collapse
Affiliation(s)
- Geula M Bernstein
- Division of Cellular and Molecular Biology, Toronto Western Research Institute, University Health Network. 399 Bathurst Street, Toronto, Ontario, Canada
| | | |
Collapse
|
47
|
Bedet C, Bruusgaard JC, Vergo S, Groth-Pedersen L, Eimer S, Triller A, Vannier C. Regulation of Gephyrin Assembly and Glycine Receptor Synaptic Stability. J Biol Chem 2006; 281:30046-56. [PMID: 16882665 DOI: 10.1074/jbc.m602155200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gephyrin is required for the formation of clusters of the glycine receptor (GlyR) in the neuronal postsynaptic membrane. It can make trimers and dimers through its N- and C-terminal G and E domains, respectively. Gephyrin oligomerization could thus create a submembrane lattice providing GlyR-binding sites. We investigated the relationships between the stability of cell surface GlyR and the ability of gephyrin splice variants to form oligomers. Using truncated and full-length gephyrins we found that the 13-amino acid sequence (cassette 5) prevents G domain trimerization. Moreover, E domain dimerization is inhibited by the gephyrin central L domain. All of the gephyrin variants bind GlyR beta subunit cytoplasmic loop with high affinity regardless of their cassette composition. Coexpression experiments in COS-7 cells demonstrated that GlyR bound to gephyrin harboring cassette 5 cannot be stabilized at the cell surface. This gephyrin variant was found to deplete synapses from both GlyR and gephyrin in transfected neurons. These data suggest that the relative expression level of cellular variants influence the overall oligomerization pattern of gephyrin and thus the turnover of synaptic GlyR.
Collapse
Affiliation(s)
- Cécile Bedet
- INSERM U789, the Laboratoire de Biologie Cellulaire de la Synapse, Ecole Normale Supérieure, F-75005, Paris, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Voltage-gated ion channels have to be at the right place in the right number to endow individual neurons with their specific character. Their biophysical properties together with their spatial distribution define the signalling characteristics of a neuron. Improper channel localization could cause communication defects in a neuronal network. This review covers recent studies of mechanisms for targeting voltage-gated ion channels to axons and dendrites, including trafficking, retention and endocytosis pathways for the preferential localization of particular ion channels. We also discuss how the spatial localization of these channels might contribute to the electrical excitability of neurons, and consider the need for future work in this emerging field.
Collapse
Affiliation(s)
- Helen C Lai
- Center for Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | |
Collapse
|
49
|
Marder E, Goaillard JM. Variability, compensation and homeostasis in neuron and network function. Nat Rev Neurosci 2006; 7:563-74. [PMID: 16791145 DOI: 10.1038/nrn1949] [Citation(s) in RCA: 755] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neurons in most animals live a very long time relative to the half-lives of all of the proteins that govern excitability and synaptic transmission. Consequently, homeostatic mechanisms are necessary to ensure stable neuronal and network function over an animal's lifetime. To understand how these homeostatic mechanisms might function, it is crucial to understand how tightly regulated synaptic and intrinsic properties must be for adequate network performance, and the extent to which compensatory mechanisms allow for multiple solutions to the production of similar behaviour. Here, we use examples from theoretical and experimental studies of invertebrates and vertebrates to explore several issues relevant to understanding the precision of tuning of synaptic and intrinsic currents for the operation of functional neuronal circuits.
Collapse
Affiliation(s)
- Eve Marder
- Volen Center and Biology Department, MS 013 Brandeis University, 415 South Street, Waltham, Massachusetts 02454, USA.
| | | |
Collapse
|
50
|
McKeown L, Robinson P, Greenwood SM, Hu W, Jones OT. PIN-G--a novel reporter for imaging and defining the effects of trafficking signals in membrane proteins. BMC Biotechnol 2006; 6:15. [PMID: 16524465 PMCID: PMC1475579 DOI: 10.1186/1472-6750-6-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 03/08/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The identification of protein trafficking signals, and their interacting mechanisms, is a fundamental objective of modern biology. Unfortunately, the analysis of trafficking signals is complicated by their topography, hierarchical nature and regulation. Powerful strategies to test candidate motifs include their ability to direct simpler reporter proteins, to which they are fused, to the appropriate cellular compartment. However, present reporters are limited by their endogenous expression, paucity of cloning sites, and difficult detection in live cells. RESULTS Consequently, we have engineered a mammalian expression vector encoding a novel trafficking reporter--pIN-G--consisting of a simple, type I integral protein bearing permissive intra/extracellular cloning sites, green fluorescent protein (GFP), cMyc and HA epitope tags. Fluorescence imaging, flow cytometry and biochemical assays of transfected HEK293 cells, confirm the size, topology and surface expression of PIN-G. Moreover, a pIN-G fusion construct, containing a Trans-Golgi Network (TGN) targeting determinant, internalises rapidly from the cell surface and localises to the TGN. Additionally, another PIN-G fusion protein and its mutants reveal trafficking determinants in the cytoplasmic carboxy terminus of Kv1.4 voltage-gated potassium channels. CONCLUSION Together, these data indicate that pIN-G is a versatile, powerful, new reporter for analysing signals controlling membrane protein trafficking, surface expression and dynamics.
Collapse
Affiliation(s)
- Lynn McKeown
- Faculty of Life Sciences, University of Manchester. 1.136 Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Philip Robinson
- Faculty of Life Sciences, University of Manchester. 1.136 Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Sam M Greenwood
- Faculty of Life Sciences, University of Manchester. 1.136 Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Weiwen Hu
- Faculty of Life Sciences, University of Manchester. 1.136 Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Owen T Jones
- Faculty of Life Sciences, University of Manchester. 1.136 Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|