1
|
Zou X, Shanmugam SK, Kanner SA, Sampson KJ, Kass RS, Colecraft HM. Divergent regulation of KCNQ1/E1 by targeted recruitment of protein kinase A to distinct sites on the channel complex. eLife 2023; 12:e83466. [PMID: 37650513 PMCID: PMC10499372 DOI: 10.7554/elife.83466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 08/30/2023] [Indexed: 09/01/2023] Open
Abstract
The slow delayed rectifier potassium current, IKs, conducted through pore-forming Q1 and auxiliary E1 ion channel complexes is important for human cardiac action potential repolarization. During exercise or fright, IKs is up-regulated by protein kinase A (PKA)-mediated Q1 phosphorylation to maintain heart rhythm and optimum cardiac performance. Sympathetic up-regulation of IKs requires recruitment of PKA holoenzyme (two regulatory - RI or RII - and two catalytic Cα subunits) to Q1 C-terminus by an A kinase anchoring protein (AKAP9). Mutations in Q1 or AKAP9 that abolish their functional interaction result in long QT syndrome type 1 and 11, respectively, which increases the risk of sudden cardiac death during exercise. Here, we investigated the utility of a targeted protein phosphorylation (TPP) approach to reconstitute PKA regulation of IKs in the absence of AKAP9. Targeted recruitment of endogenous Cα to E1-YFP using a GFP/YFP nanobody (nano) fused to RIIα enabled acute cAMP-mediated enhancement of IKs, reconstituting physiological regulation of the channel complex. By contrast, nano-mediated tethering of RIIα or Cα to Q1-YFP constitutively inhibited IKs by retaining the channel intracellularly in the endoplasmic reticulum and Golgi. Proteomic analysis revealed that distinct phosphorylation sites are modified by Cα targeted to Q1-YFP compared to free Cα. Thus, functional outcomes of synthetically recruited PKA on IKs regulation is critically dependent on the site of recruitment within the channel complex. The results reveal insights into divergent regulation of IKs by phosphorylation across different spatial and time scales, and suggest a TPP approach to develop new drugs to prevent exercise-induced sudden cardiac death.
Collapse
Affiliation(s)
- Xinle Zou
- Department of Molecular Pharmacology and Therapeutics, Columbia UniversityNew YorkUnited States
| | - Sri Karthika Shanmugam
- Department of Physiology and Cellular Biophysics, Columbia UniversityNew YorkUnited States
| | - Scott A Kanner
- Doctoral Program in Neurobiology and Behavior, Columbia UniversityNew YorkUnited States
| | - Kevin J Sampson
- Department of Molecular Pharmacology and Therapeutics, Columbia UniversityNew YorkUnited States
| | - Robert S Kass
- Department of Molecular Pharmacology and Therapeutics, Columbia UniversityNew YorkUnited States
| | - Henry M Colecraft
- Department of Molecular Pharmacology and Therapeutics, Columbia UniversityNew YorkUnited States
- Doctoral Program in Neurobiology and Behavior, Columbia UniversityNew YorkUnited States
| |
Collapse
|
2
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
3
|
GAD67-mediated GABA Synthesis and Signaling Impinges on Directing Basket Cell Axonal Projections Toward Purkinje Cells in the Cerebellum. THE CEREBELLUM 2021; 21:905-919. [PMID: 34676525 DOI: 10.1007/s12311-021-01334-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
Gamma-aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the central nervous system, synthesized by two isoforms of glutamate decarboxylase (GAD): GAD65 and GAD67. GABA may act as a trophic factor during brain development, but its contribution to the development and maturation of cerebellar neural circuits is not known. To understand the roles of GABA in cerebellar organization and associated functions in motor coordination and balance, we examined GAD65 conventional knock out (KO) mice and mice in which GAD67 was eliminated in parvalbumin-expressing neurons (PV-Cre; GAD67flox/flox mice). We found aberrant subcellular localization of the Shaker-type K channel Kv1.1 in basket cell collaterals of PV-Cre; GAD67 flox/flox mice and abnormal projections from basket cells to Purkinje cells in both mouse strains. We also found that altered synaptic properties of basket cell terminals to Purkinje cells in PV-Cre; GAD67flox/flox mice. Furthermore, PV-Cre; GAD67 flox/flox mice exhibited abnormal motor coordination in the rotarod test. These results indicate that GABA signaling in the cerebellum is critical for establishing appropriate connections between basket cells and Purkinje cells and is associated with motor coordination in mice.
Collapse
|
4
|
Abstract
K+ channels enable potassium to flow across the membrane with great selectivity. There are four K+ channel families: voltage-gated K (Kv), calcium-activated (KCa), inwardly rectifying K (Kir), and two-pore domain potassium (K2P) channels. All four K+ channels are formed by subunits assembling into a classic tetrameric (4x1P = 4P for the Kv, KCa, and Kir channels) or tetramer-like (2x2P = 4P for the K2P channels) architecture. These subunits can either be the same (homomers) or different (heteromers), conferring great diversity to these channels. They share a highly conserved selectivity filter within the pore but show different gating mechanisms adapted for their function. K+ channels play essential roles in controlling neuronal excitability by shaping action potentials, influencing the resting membrane potential, and responding to diverse physicochemical stimuli, such as a voltage change (Kv), intracellular calcium oscillations (KCa), cellular mediators (Kir), or temperature (K2P).
Collapse
|
5
|
Wu J, Kaczmarek LK. Modulation of Neuronal Potassium Channels During Auditory Processing. Front Neurosci 2021; 15:596478. [PMID: 33613177 PMCID: PMC7887315 DOI: 10.3389/fnins.2021.596478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/11/2021] [Indexed: 11/16/2022] Open
Abstract
The extraction and localization of an auditory stimulus of interest from among multiple other sounds, as in the ‘cocktail-party’ situation, requires neurons in auditory brainstem nuclei to encode the timing, frequency, and intensity of sounds with high fidelity, and to compare inputs coming from the two cochleae. Accurate localization of sounds requires certain neurons to fire at high rates with high temporal accuracy, a process that depends heavily on their intrinsic electrical properties. Studies have shown that the membrane properties of auditory brainstem neurons, particularly their potassium currents, are not fixed but are modulated in response to changes in the auditory environment. Here, we review work focusing on how such modulation of potassium channels is critical to shaping the firing pattern and accuracy of these neurons. We describe how insights into the role of specific channels have come from human gene mutations that impair localization of sounds in space. We also review how short-term and long-term modulation of these channels maximizes the extraction of auditory information, and how errors in the regulation of these channels contribute to deficits in decoding complex auditory information.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
6
|
Smith PA. K + Channels in Primary Afferents and Their Role in Nerve Injury-Induced Pain. Front Cell Neurosci 2020; 14:566418. [PMID: 33093824 PMCID: PMC7528628 DOI: 10.3389/fncel.2020.566418] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Sensory abnormalities generated by nerve injury, peripheral neuropathy or disease are often expressed as neuropathic pain. This type of pain is frequently resistant to therapeutic intervention and may be intractable. Numerous studies have revealed the importance of enduring increases in primary afferent excitability and persistent spontaneous activity in the onset and maintenance of peripherally induced neuropathic pain. Some of this activity results from modulation, increased activity and /or expression of voltage-gated Na+ channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. K+ channels expressed in dorsal root ganglia (DRG) include delayed rectifiers (Kv1.1, 1.2), A-channels (Kv1.4, 3.3, 3.4, 4.1, 4.2, and 4.3), KCNQ or M-channels (Kv7.2, 7.3, 7.4, and 7.5), ATP-sensitive channels (KIR6.2), Ca2+-activated K+ channels (KCa1.1, 2.1, 2.2, 2.3, and 3.1), Na+-activated K+ channels (KCa4.1 and 4.2) and two pore domain leak channels (K2p; TWIK related channels). Function of all K+ channel types is reduced via a multiplicity of processes leading to altered expression and/or post-translational modification. This also increases excitability of DRG cell bodies and nociceptive free nerve endings, alters axonal conduction and increases neurotransmitter release from primary afferent terminals in the spinal dorsal horn. Correlation of these cellular changes with behavioral studies provides almost indisputable evidence for K+ channel dysfunction in the onset and maintenance of neuropathic pain. This idea is underlined by the observation that selective impairment of just one subtype of DRG K+ channel can produce signs of pain in vivo. Whilst it is established that various mediators, including cytokines and growth factors bring about injury-induced changes in DRG function and excitability, evidence presently available points to a seminal role for interleukin 1β (IL-1β) in control of K+ channel function. Despite the current state of knowledge, attempts to target K+ channels for therapeutic pain management have met with limited success. This situation may change with the advent of personalized medicine. Identification of specific sensory abnormalities and genetic profiling of individual patients may predict therapeutic benefit of K+ channel activators.
Collapse
Affiliation(s)
- Peter A. Smith
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
7
|
Chao OY, Marron Fernandez de Velasco E, Pathak SS, Maitra S, Zhang H, Duvick L, Wickman K, Orr HT, Hirai H, Yang YM. Targeting inhibitory cerebellar circuitry to alleviate behavioral deficits in a mouse model for studying idiopathic autism. Neuropsychopharmacology 2020; 45:1159-1170. [PMID: 32179875 PMCID: PMC7234983 DOI: 10.1038/s41386-020-0656-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/11/2020] [Accepted: 03/03/2020] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) encompasses wide-ranging neuropsychiatric symptoms with unclear etiology. Although the cerebellum is a key region implicated in ASD, it remains elusive how the cerebellar circuitry is altered and whether the cerebellum can serve as a therapeutic target to rectify the phenotype of idiopathic ASD with polygenic abnormalities. Using a syndromic ASD model, e.g., Black and Tan BRachyury T+Itpr3tf/J (BTBR) mice, we revealed that increased excitability of presynaptic interneurons (INs) and decreased intrinsic excitability of postsynaptic Purkinje neurons (PNs) resulted in low PN firing rates in the cerebellum. Knowing that downregulation of Kv1.2 potassium channel in the IN nerve terminals likely augmented their excitability and GABA release, we applied a positive Kv1.2 modulator to mitigate the presynaptic over-inhibition and social impairment of BTBR mice. Selective restoration of the PN activity by a new chemogenetic approach alleviated core ASD-like behaviors of the BTBR strain. These findings highlight complex mechanisms converging onto the cerebellar dysfunction in the phenotypic model and provide effective strategies for potential therapies of ASD.
Collapse
Affiliation(s)
- Owen Y Chao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | | | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Swati Maitra
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Hao Zhang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA.
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
8
|
Identification of a molecular locus for normalizing dysregulated GABA release from interneurons in the Fragile X brain. Mol Psychiatry 2020; 25:2017-2035. [PMID: 30224722 PMCID: PMC7473840 DOI: 10.1038/s41380-018-0240-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/07/2018] [Accepted: 08/10/2018] [Indexed: 01/14/2023]
Abstract
Principal neurons encode information by varying their firing rate and patterns precisely fine-tuned through GABAergic interneurons. Dysregulation of inhibition can lead to neuropsychiatric disorders, yet little is known about the molecular basis underlying inhibitory control. Here, we find that excessive GABA release from basket cells (BCs) attenuates the firing frequency of Purkinje neurons (PNs) in the cerebellum of Fragile X Mental Retardation 1 (Fmr1) knockout (KO) mice, a model of Fragile X Syndrome (FXS) with abrogated expression of the Fragile X Mental Retardation Protein (FMRP). This over-inhibition originates from increased excitability and Ca2+ transients in the presynaptic terminals, where Kv1.2 potassium channels are downregulated. By paired patch-clamp recordings, we further demonstrate that acutely introducing an N-terminal fragment of FMRP into BCs normalizes GABA release in the Fmr1-KO synapses. Conversely, direct injection of an inhibitory FMRP antibody into BCs, or membrane depolarization of BCs, enhances GABA release in the wild type synapses, leading to abnormal inhibitory transmission comparable to the Fmr1-KO neurons. We discover that the N-terminus of FMRP directly binds to a phosphorylated serine motif on the C-terminus of Kv1.2; and that loss of this interaction in BCs exaggerates GABA release, compromising the firing activity of PNs and thus the output from the cerebellar circuitry. An allosteric Kv1.2 agonist, docosahexaenoic acid, rectifies the dysregulated inhibition in vitro as well as acoustic startle reflex and social interaction in vivo of the Fmr1-KO mice. Our results unravel a novel molecular locus for targeted intervention of FXS and perhaps autism.
Collapse
|
9
|
Capera J, Serrano-Novillo C, Navarro-Pérez M, Cassinelli S, Felipe A. The Potassium Channel Odyssey: Mechanisms of Traffic and Membrane Arrangement. Int J Mol Sci 2019; 20:ijms20030734. [PMID: 30744118 PMCID: PMC6386995 DOI: 10.3390/ijms20030734] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/29/2022] Open
Abstract
Ion channels are transmembrane proteins that conduct specific ions across biological membranes. Ion channels are present at the onset of many cellular processes, and their malfunction triggers severe pathologies. Potassium channels (KChs) share a highly conserved signature that is necessary to conduct K⁺ through the pore region. To be functional, KChs require an exquisite regulation of their subcellular location and abundance. A wide repertoire of signatures facilitates the proper targeting of the channel, fine-tuning the balance that determines traffic and location. These signature motifs can be part of the secondary or tertiary structure of the protein and are spread throughout the entire sequence. Furthermore, the association of the pore-forming subunits with different ancillary proteins forms functional complexes. These partners can modulate traffic and activity by adding their own signatures as well as by exposing or masking the existing ones. Post-translational modifications (PTMs) add a further dimension to traffic regulation. Therefore, the fate of a KCh is not fully dependent on a gene sequence but on the balance of many other factors regulating traffic. In this review, we assemble recent evidence contributing to our understanding of the spatial expression of KChs in mammalian cells. We compile specific signatures, PTMs, and associations that govern the destination of a functional channel.
Collapse
Affiliation(s)
- Jesusa Capera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Clara Serrano-Novillo
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - María Navarro-Pérez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Silvia Cassinelli
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Avda. Diagonal 643, 08028 Barcelona, Spain.
| |
Collapse
|
10
|
Yang JW, Larson G, Konrad L, Shetty M, Holy M, Jäntsch K, Kastein M, Heo S, Erdem FA, Lubec G, Vaughan RA, Sitte HH, Foster JD. Dephosphorylation of human dopamine transporter at threonine 48 by protein phosphatase PP1/2A up-regulates transport velocity. J Biol Chem 2018; 294:3419-3431. [PMID: 30587577 DOI: 10.1074/jbc.ra118.005251] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/20/2018] [Indexed: 11/06/2022] Open
Abstract
Several protein kinases, including protein kinase C, Ca2+/calmodulin-dependent protein kinase II, and extracellular signal-regulated kinase, play key roles in the regulation of dopamine transporter (DAT) functions. These functions include surface expression, internalization, and forward and reverse transport, with phosphorylation sites for these kinases being linked to distinct regions of the DAT N terminus. Protein phosphatases (PPs) also regulate DAT activity, but the specific residues associated with their activities have not yet been elucidated. In this study, using co-immunoprecipitation followed by MS and immunoblotting analyses, we demonstrate the association of DAT with PP1 and PP2A in the mouse brain and heterologous cell systems. By applying MS in conjunction with a metabolic labeling method, we defined a PP1/2A-sensitive phosphorylation site at Thr-48 in human DAT, a residue that has not been previously reported to be involved in DAT phosphorylation. Site-directed mutagenesis of Thr-48 to Ala (T48A) to prevent phosphorylation enhanced dopamine transport kinetics, supporting a role for this residue in regulating DAT activity. Moreover, T48A-DAT displayed increased palmitoylation, suggesting that phosphorylation/dephosphorylation at this site has an additional regulatory role and reinforcing a previously reported reciprocal relationship between C-terminal palmitoylation and N-terminal phosphorylation.
Collapse
Affiliation(s)
- Jae-Won Yang
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Garret Larson
- the Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9037, and
| | - Lisa Konrad
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Madhur Shetty
- the Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9037, and
| | - Marion Holy
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Kathrin Jäntsch
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Mirja Kastein
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Seok Heo
- the Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Fatma Asli Erdem
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - Gert Lubec
- Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Roxanne A Vaughan
- the Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9037, and
| | - Harald H Sitte
- From the Institute of Pharmacology, Center for Physiology and Pharmacology, and
| | - James D Foster
- the Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202-9037, and
| |
Collapse
|
11
|
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: 0.7] [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
|
12
|
Masnada S, Hedrich UBS, Gardella E, Schubert J, Kaiwar C, Klee EW, Lanpher BC, Gavrilova RH, Synofzik M, Bast T, Gorman K, King MD, Allen NM, Conroy J, Ben Zeev B, Tzadok M, Korff C, Dubois F, Ramsey K, Narayanan V, Serratosa JM, Giraldez BG, Helbig I, Marsh E, O'Brien M, Bergqvist CA, Binelli A, Porter B, Zaeyen E, Horovitz DD, Wolff M, Marjanovic D, Caglayan HS, Arslan M, Pena SDJ, Sisodiya SM, Balestrini S, Syrbe S, Veggiotti P, Lemke JR, Møller RS, Lerche H, Rubboli G. Clinical spectrum and genotype-phenotype associations of KCNA2-related encephalopathies. Brain 2017; 140:2337-2354. [PMID: 29050392 DOI: 10.1093/brain/awx184] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 06/06/2017] [Indexed: 11/14/2022] Open
Abstract
Recently, de novo mutations in the gene KCNA2, causing either a dominant-negative loss-of-function or a gain-of-function of the voltage-gated K+ channel Kv1.2, were described to cause a new molecular entity within the epileptic encephalopathies. Here, we report a cohort of 23 patients (eight previously described) with epileptic encephalopathy carrying either novel or known KCNA2 mutations, with the aim to detail the clinical phenotype associated with each of them, to characterize the functional effects of the newly identified mutations, and to assess genotype-phenotype associations. We identified five novel and confirmed six known mutations, three of which recurred in three, five and seven patients, respectively. Ten mutations were missense and one was a truncation mutation; de novo occurrence could be shown in 20 patients. Functional studies using a Xenopus oocyte two-microelectrode voltage clamp system revealed mutations with only loss-of-function effects (mostly dominant-negative current amplitude reduction) in eight patients or only gain-of-function effects (hyperpolarizing shift of voltage-dependent activation, increased amplitude) in nine patients. In six patients, the gain-of-function was diminished by an additional loss-of-function (gain-and loss-of-function) due to a hyperpolarizing shift of voltage-dependent activation combined with either decreased amplitudes or an additional hyperpolarizing shift of the inactivation curve. These electrophysiological findings correlated with distinct phenotypic features. The main differences were (i) predominant focal (loss-of-function) versus generalized (gain-of-function) seizures and corresponding epileptic discharges with prominent sleep activation in most cases with loss-of-function mutations; (ii) more severe epilepsy, developmental problems and ataxia, and atrophy of the cerebellum or even the whole brain in about half of the patients with gain-of-function mutations; and (iii) most severe early-onset phenotypes, occasionally with neonatal onset epilepsy and developmental impairment, as well as generalized and focal seizures and EEG abnormalities for patients with gain- and loss-of-function mutations. Our study thus indicates well represented genotype-phenotype associations between three subgroups of patients with KCNA2 encephalopathy according to the electrophysiological features of the mutations.
Collapse
Affiliation(s)
- Silvia Masnada
- Brain and Behaviour Department, University of Pavia, Italy.,Department of Child Neurology and Psychiatry, IRCCS C. Mondino National Neurological Institute, Pavia, Italy.,Danish Epilepsy Centre, Dianalund, Denmark
| | - Ulrike B S Hedrich
- Department of Neurology and Epileptology, Hertie Institute of Clinical Brain Research, University of Tübingen, Germany
| | - Elena Gardella
- Clinical Neurophysiology, Danish Epilepsy Centre, Dianalund, Denmark.,Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark
| | - Julian Schubert
- Department of Neurology and Epileptology, Hertie Institute of Clinical Brain Research, University of Tübingen, Germany
| | - Charu Kaiwar
- Center for Individualized Medicine, Mayo Clinic, Scottsdale AZ, USA
| | - Eric W Klee
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Ralitza H Gavrilova
- Departments of Clinical Genomics and Neurology Mayo Clinic, Rochester, MN, USA
| | - Matthis Synofzik
- Department of Neurodegeneration, Hertie Institute for Clinical Brain Research (HIH), University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Kathleen Gorman
- Department of Neurology and Clinical Neurophysiology, Children's University Hospital, Temple Street, Dublin, Ireland.,Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Ireland
| | - Mary D King
- Department of Neurology and Clinical Neurophysiology, Children's University Hospital, Temple Street, Dublin, Ireland.,Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Ireland
| | - Nicholas M Allen
- Department of Neurology and Clinical Neurophysiology, Children's University Hospital, Temple Street, Dublin, Ireland.,Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Ireland
| | - Judith Conroy
- Academic Centre on Rare Diseases, School of Medicine and Medical Science, University College Dublin, Ireland
| | - Bruria Ben Zeev
- Sackler school of medicine Tel Aviv University, Tel Aviv, Israel
| | - Michal Tzadok
- Pediatric Neurology Unit, Sheba Medical Center, Israel
| | - Christian Korff
- Pediatric Neurology University Hospitals, Geneva, Switzerland
| | | | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA.,Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, AZ, USA.,Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Jose M Serratosa
- Neurology Laboratory and Epilepsy Unit, Department of Neurology, IIS- Fundación Jiménez Díaz, UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Beatriz G Giraldez
- Neurology Laboratory and Epilepsy Unit, Department of Neurology, IIS- Fundación Jiménez Díaz, UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ingo Helbig
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, USA.,Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Kiel Germany
| | - Eric Marsh
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Margaret O'Brien
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, USA
| | | | - Adrian Binelli
- Department of Pediatric Neurology, Elizalde Children's Hospital, Buenos Aires, Argentina.,Sociedad Argentina de Neurología Infantil (SANI) / Argentinian Child Neurology Society
| | - Brenda Porter
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto California, USA
| | - Eduardo Zaeyen
- Neuropediatrics Committee of State of Rio De Janeiro, Rio De Janeiro, Brazil
| | - Dafne D Horovitz
- Medical Genetics Department, National Institute for Women, Children and Adolescents Health Fernandes Figueira - Fiocruz, Rio de Janeiro, Brazil
| | - Markus Wolff
- Department of Pediatric Neurology and Developmental Medecine, University Children's Hospital, Tübingen, Germany
| | | | - Hande S Caglayan
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey
| | - Mutluay Arslan
- Department of Pediatric Neurology, Gulhane Military Medical School, Ankara, Turkey
| | - Sergio D J Pena
- GENE - Núcleo de Genética Médica, Belo Horizonte, MG, Brazil
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and Epilepsy Society, UK
| | - Simona Balestrini
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and Epilepsy Society, UK
| | - Steffen Syrbe
- Department of General Paediatrics, Division of Child Neurology and Inherited Metabolic Diseases, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig Hospitals and Clinics, Leipzig, Germany
| | - Pierangelo Veggiotti
- Brain and Behaviour Department, University of Pavia, Italy.,Department of Child Neurology and Psychiatry, IRCCS C. Mondino National Neurological Institute, Pavia, Italy
| | | | - Rikke S Møller
- Danish Epilepsy Centre, Dianalund, Denmark.,Institute for Regional Health Services, University of Southern Denmark, Odense, Denmark
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute of Clinical Brain Research, University of Tübingen, Germany
| | - Guido Rubboli
- Danish Epilepsy Centre, Dianalund, Denmark.,University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Nongenomic Glucocorticoid Suppression of a Postsynaptic Potassium Current via Emergent Autocrine Endocannabinoid Signaling in Hypothalamic Neuroendocrine Cells following Chronic Dehydration. eNeuro 2017; 4:eN-NWR-0216-17. [PMID: 28966975 PMCID: PMC5617081 DOI: 10.1523/eneuro.0216-17.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/19/2017] [Accepted: 08/22/2017] [Indexed: 11/21/2022] Open
Abstract
Glucocorticoids rapidly stimulate endocannabinoid synthesis and modulation of synaptic transmission in hypothalamic neuroendocrine cells via a nongenomic signaling mechanism. The endocannabinoid actions are synapse-constrained by astrocyte restriction of extracellular spatial domains. Exogenous cannabinoids have been shown to modulate postsynaptic potassium currents, including the A-type potassium current (IA), in different cell types. The activity of magnocellular neuroendocrine cells is shaped by a prominent IA. We tested for a rapid glucocorticoid modulation of the postsynaptic IK and IA in magnocellular neuroendocrine cells of the hypothalamic paraventricular nucleus (PVN) using whole-cell recordings in rat brain slices. Application of the synthetic glucocorticoid dexamethasone (Dex) had no rapid effect on the IK or IA amplitude, voltage dependence, or kinetics in magnocellular neurons in slices from untreated rats. In magnocellular neurons from salt-loaded rats, however, Dex application caused a rapid suppression of the IA and a depolarizing shift in IA voltage dependence. Exogenously applied endocannabinoids mimicked the rapid Dex modulation of the IA, and CB1 receptor antagonists and agonists blocked and occluded the Dex-induced changes in the IA, respectively, suggesting an endocannabinoid dependence of the rapid glucocorticoid effect. Preincubation of control slices in a gliotoxin resulted in the partial recapitulation of the glucocorticoid-induced rapid suppression of the IA. These findings demonstrate a glucocorticoid suppression of the postsynaptic IA in PVN magnocellular neurons via an autocrine endocannabinoid-dependent mechanism following chronic dehydration, and suggest a possible role for astrocytes in the control of the autocrine endocannabinoid actions.
Collapse
|
14
|
Vicente PC, Kim JY, Ha J, Song M, Lee H, Kim D, Choi J, Park K. Identification and characterization of site‐specific N‐glycosylation in the potassium channel Kv3.1b. J Cell Physiol 2017; 233:549-558. [DOI: 10.1002/jcp.25915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 03/17/2017] [Indexed: 12/11/2022]
Affiliation(s)
| | - Jin Young Kim
- Biomedical Omics GroupKorea Basic Science InstituteCheongju‐si Chungcheongbuk‐doSouth Korea
| | - Jeong‐Ju Ha
- Department of Physiology, School of MedicineKyung Hee UniversitySeoulSouth Korea
| | - Min‐Young Song
- Department of Physiology, School of MedicineKyung Hee UniversitySeoulSouth Korea
- Biomedical Omics GroupKorea Basic Science InstituteCheongju‐si Chungcheongbuk‐doSouth Korea
| | - Hyun‐Kyung Lee
- Biomedical Omics GroupKorea Basic Science InstituteCheongju‐si Chungcheongbuk‐doSouth Korea
- Graduate School of Analytical Science and TechnologyChungnam National UniversityDaejeonSouth Korea
| | - Dong‐Hyun Kim
- College of PharmacyCatholic University of KoreaBucheonGyeonggi‐DoSouth Korea
| | - Jin‐Sung Choi
- College of PharmacyCatholic University of KoreaBucheonGyeonggi‐DoSouth Korea
| | - Kang‐Sik Park
- Department of Physiology, School of MedicineKyung Hee UniversitySeoulSouth Korea
| |
Collapse
|
15
|
Salzer I, Erdem FA, Chen WQ, Heo S, Koenig X, Schicker KW, Kubista H, Lubec G, Boehm S, Yang JW. Phosphorylation regulates the sensitivity of voltage-gated Kv7.2 channels towards phosphatidylinositol-4,5-bisphosphate. J Physiol 2016; 595:759-776. [PMID: 27621207 PMCID: PMC5215842 DOI: 10.1113/jp273274] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/30/2016] [Indexed: 12/22/2022] Open
Abstract
Key points Phosphatidylinositol‐4,5‐bisphosphate (PIP2) is a key regulator of many membrane proteins, including voltage‐gated Kv7.2 channels. In this study, we identified the residues in five phosphorylation sites and their corresponding protein kinases, the former being clustered within one of four putative PIP2‐binding domains in Kv7.2. Dephosphorylation of these residues reduced the sensitivity of Kv7.2 channels towards PIP2. Dephosphorylation of Kv7.2 affected channel inhibition via M1 muscarinic receptors, but not via bradykinin receptors. Our data indicated that phosphorylation of the Kv7.2 channel was necessary to maintain its low affinity for PIP2, thereby ensuring the tight regulation of the channel via G protein‐coupled receptors.
Abstract The function of numerous ion channels is tightly controlled by G protein‐coupled receptors (GPCRs). The underlying signalling mechanisms may involve phosphorylation of channel proteins and participation of phosphatidylinositol‐4,5‐bisphosphate (PIP2). Although the roles of both mechanisms have been investigated extensively, thus far only little has been reported on their interaction in channel modulation. GPCRs govern Kv7 channels, the latter playing a major role in the regulation of neuronal excitability by determining the levels of PIP2 and through phosphorylation. Using liquid chromatography‐coupled mass spectrometry for Kv7.2 immunoprecipitates of rat brain membranes and transfected cells, we mapped a cluster of five phosphorylation sites in one of the PIP2‐binding domains. To evaluate the effect of phosphorylation on PIP2‐mediated Kv7.2 channel regulation, a quintuple alanine mutant of these serines (S427/S436/S438/S446/S455; A5 mutant) was generated to mimic the dephosphorylated state. Currents passing through these mutated channels were less sensitive towards PIP2 depletion via the voltage‐sensitive phosphatase Dr‐VSP than were wild‐type channels. In vitro phosphorylation assays with the purified C‐terminus of Kv7.2 revealed that CDK5, p38 MAPK, CaMKIIα and PKA were able to phosphorylate the five serines. Inhibition of these protein kinases reduced the sensitivity of wild‐type but not mutant Kv7.2 channels towards PIP2 depletion via Dr‐VSP. In superior cervical ganglion neurons, the protein kinase inhibitors attenuated Kv7 current regulation via M1 receptors, but left unaltered the control by B2 receptors. Our results revealed that the phosphorylation status of serines located within a putative PIP2‐binding domain determined the phospholipid sensitivity of Kv7.2 channels and supported GPCR‐mediated channel regulation. Phosphatidylinositol‐4,5‐bisphosphate (PIP2) is a key regulator of many membrane proteins, including voltage‐gated Kv7.2 channels. In this study, we identified the residues in five phosphorylation sites and their corresponding protein kinases, the former being clustered within one of four putative PIP2‐binding domains in Kv7.2. Dephosphorylation of these residues reduced the sensitivity of Kv7.2 channels towards PIP2. Dephosphorylation of Kv7.2 affected channel inhibition via M1 muscarinic receptors, but not via bradykinin receptors. Our data indicated that phosphorylation of the Kv7.2 channel was necessary to maintain its low affinity for PIP2, thereby ensuring the tight regulation of the channel via G protein‐coupled receptors.
Collapse
Affiliation(s)
- Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Fatma Asli Erdem
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Wei-Qiang Chen
- Department of Pediatrics, Medical University of Vienna, 1090, Vienna, Austria
| | - Seok Heo
- Department of Pediatrics, Medical University of Vienna, 1090, Vienna, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Klaus W Schicker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Gert Lubec
- Department of Pharmaceutical Chemistry, University of Vienna, 1090, Vienna, Austria
| | - Stefan Boehm
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
16
|
Dissecting the Role of P/Q-Type Calcium Channels in Corticothalamic Circuit Dysfunction and Absence Epilepsy. J Neurosci 2016; 36:5677-9. [PMID: 27225758 DOI: 10.1523/jneurosci.0753-16.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022] Open
|
17
|
Calvo M, Richards N, Schmid AB, Barroso A, Zhu L, Ivulic D, Zhu N, Anwandter P, Bhat MA, Court FA, McMahon SB, Bennett DLH. Altered potassium channel distribution and composition in myelinated axons suppresses hyperexcitability following injury. eLife 2016; 5:e12661. [PMID: 27033551 PMCID: PMC4841771 DOI: 10.7554/elife.12661] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/15/2016] [Indexed: 12/25/2022] Open
Abstract
Neuropathic pain following peripheral nerve injury is associated with hyperexcitability in damaged myelinated sensory axons, which begins to normalise over time. We investigated the composition and distribution of shaker-type-potassium channels (Kv1 channels) within the nodal complex of myelinated axons following injury. At the neuroma that forms after damage, expression of Kv1.1 and 1.2 (normally localised to the juxtaparanode) was markedly decreased. In contrast Kv1.4 and 1.6, which were hardly detectable in the naïve state, showed increased expression within juxtaparanodes and paranodes following injury, both in rats and humans. Within the dorsal root (a site remote from injury) we noted a redistribution of Kv1-channels towards the paranode. Blockade of Kv1 channels with α-DTX after injury reinstated hyperexcitability of A-fibre axons and enhanced mechanosensitivity. Changes in the molecular composition and distribution of axonal Kv1 channels, therefore represents a protective mechanism to suppress the hyperexcitability of myelinated sensory axons that follows nerve injury.
Collapse
Affiliation(s)
- Margarita Calvo
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile.,Departamento de Anestesiologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Natalie Richards
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - Annina B Schmid
- School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane, Australia.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Alejandro Barroso
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,Hospital Regional Universitario de Málaga. Servicio de Anestesiología, Málaga, Spain
| | - Lan Zhu
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom.,School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Dinka Ivulic
- Departamento de Fisiologia, Facultad de Ciencias Biologicas- Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ning Zhu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Philipp Anwandter
- Departamento Ortopedia y Traumatologia, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Manzoor A Bhat
- Department of Physiology, UT Health Science Center at San Antonio, San Antonio, United States.,School of Medicine, UT Health Science Center at San Antonio, San Antonio, United States
| | - Felipe A Court
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile.,FONDAP, Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Millenium Nucleus for Regenerative Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Kings College London, London, United Kingdom
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Baronas VA, Yang R, Vilin YY, Kurata HT. Determinants of frequency-dependent regulation of Kv1.2-containing potassium channels. Channels (Austin) 2015; 10:158-66. [PMID: 26646078 PMCID: PMC4960988 DOI: 10.1080/19336950.2015.1120390] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated potassium channels are important regulators of electrical excitation in many tissues, with Kv1.2 standing out as an essential contributor in the CNS. Genetic deletion of Kv1.2 invariably leads to early lethality in mice. In humans, mutations affecting Kv1.2 function are linked to epileptic encephalopathy and movement disorders. We have demonstrated that Kv1.2 is subject to a unique regulatory mechanism in which repetitive stimulation leads to dramatic potentiation of current. In this study, we explore the properties and molecular determinants of this use-dependent potentiation/activation. First, we examine how alterations in duty cycle (depolarization and repolarization/recovery times) affect the onset and extent of use-dependent activation. Also, we use trains of repetitive depolarizations to test the effects of a variety of Thr252 (S2-S3 linker) mutations on use-dependent activation. Substitutions of Thr with some sterically similar amino acids (Ser, Val, and Met, but not Cys) retain use-dependent activation, while bulky or charged amino acid substitutions eliminate use-dependence. Introduction of Thr at the equivalent position in other Kv1 channels (1.1, 1.3, 1.4), was not sufficient to transfer the phenotype. We hypothesize that use-dependent activation of Kv1.2 channels is mediated by an extrinsic regulator that binds preferentially to the channel closed state, with Thr252 being necessary but not sufficient for this interaction to alter channel function. These findings extend the conclusions of our recent demonstration of use-dependent activation of Kv1.2-containing channels in hippocampal neurons, by adding new details about the molecular mechanism underlying this effect.
Collapse
Affiliation(s)
- Victoria A Baronas
- a Anesthesiology, Pharmacology, and Therapeutics , University of British Columbia , Vancouver , BC , Canada
| | - Runying Yang
- b Department of Pharmacology , University of Alberta , Edmonton , AB , Canada
| | - Yury Y Vilin
- a Anesthesiology, Pharmacology, and Therapeutics , University of British Columbia , Vancouver , BC , Canada
| | - Harley T Kurata
- b Department of Pharmacology , University of Alberta , Edmonton , AB , Canada
| |
Collapse
|
19
|
Phosphorylation of the Cav3.2 T-type calcium channel directly regulates its gating properties. Proc Natl Acad Sci U S A 2015; 112:13705-10. [PMID: 26483470 DOI: 10.1073/pnas.1511740112] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Phosphorylation is a major mechanism regulating the activity of ion channels that remains poorly understood with respect to T-type calcium channels (Cav3). These channels are low voltage-activated calcium channels that play a key role in cellular excitability and various physiological functions. Their dysfunction has been linked to several neurological disorders, including absence epilepsy and neuropathic pain. Recent studies have revealed that T-type channels are modulated by a variety of serine/threonine protein kinase pathways, which indicates the need for a systematic analysis of T-type channel phosphorylation. Here, we immunopurified Cav3.2 channels from rat brain, and we used high-resolution MS to construct the first, to our knowledge, in vivo phosphorylation map of a voltage-gated calcium channel in a mammalian brain. We identified as many as 34 phosphorylation sites, and we show that the vast majority of these sites are also phosphorylated on the human Cav3.2 expressed in HEK293T cells. In patch-clamp studies, treatment of the channel with alkaline phosphatase as well as analysis of dephosphomimetic mutants revealed that phosphorylation regulates important functional properties of Cav3.2 channels, including voltage-dependent activation and inactivation and kinetics. We also identified that the phosphorylation of a locus situated in the loop I-II S442/S445/T446 is crucial for this regulation. Our data show that Cav3.2 channels are highly phosphorylated in the mammalian brain and establish phosphorylation as an important mechanism involved in the dynamic regulation of Cav3.2 channel gating properties.
Collapse
|
20
|
Trimmer JS. Subcellular localization of K+ channels in mammalian brain neurons: remarkable precision in the midst of extraordinary complexity. Neuron 2015; 85:238-56. [PMID: 25611506 DOI: 10.1016/j.neuron.2014.12.042] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Potassium channels (KChs) are the most diverse ion channels, in part due to extensive combinatorial assembly of a large number of principal and auxiliary subunits into an assortment of KCh complexes. Their structural and functional diversity allows KChs to play diverse roles in neuronal function. Localization of KChs within specialized neuronal compartments defines their physiological role and also fundamentally impacts their activity, due to localized exposure to diverse cellular determinants of channel function. Recent studies in mammalian brain reveal an exquisite refinement of KCh subcellular localization. This includes axonal KChs at the initial segment, and near/within nodes of Ranvier and presynaptic terminals, dendritic KChs found at sites reflecting specific synaptic input, and KChs defining novel neuronal compartments. Painting the remarkable diversity of KChs onto the complex architecture of mammalian neurons creates an elegant picture of electrical signal processing underlying the sophisticated function of individual neuronal compartments, and ultimately neurotransmission and behavior.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
21
|
Regulation of the cardiac Na+ channel NaV1.5 by post-translational modifications. J Mol Cell Cardiol 2015; 82:36-47. [PMID: 25748040 DOI: 10.1016/j.yjmcc.2015.02.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/28/2015] [Accepted: 02/17/2015] [Indexed: 02/07/2023]
Abstract
The cardiac voltage-gated Na(+) channel, Na(V)1.5, is responsible for the upstroke of the action potential in cardiomyocytes and for efficient propagation of the electrical impulse in the myocardium. Even subtle alterations of Na(V)1.5 function, as caused by mutations in its gene SCN5A, may lead to many different arrhythmic phenotypes in carrier patients. In addition, acquired malfunctions of Na(V)1.5 that are secondary to cardiac disorders such as heart failure and cardiomyopathies, may also play significant roles in arrhythmogenesis. While it is clear that the regulation of Na(V)1.5 protein expression and function tightly depends on genetic mechanisms, recent studies have demonstrated that Na(V)1.5 is the target of various post-translational modifications that are pivotal not only in physiological conditions, but also in disease. In this review, we examine the recent literature demonstrating glycosylation, phosphorylation by Protein Kinases A and C, Ca(2+)/Calmodulin-dependent protein Kinase II, Phosphatidylinositol 3-Kinase, Serum- and Glucocorticoid-inducible Kinases, Fyn and Adenosine Monophosphate-activated Protein Kinase, methylation, acetylation, redox modifications, and ubiquitylation of Na(V)1.5. Modern and sensitive mass spectrometry approaches, applied directly to channel proteins that were purified from native cardiac tissues, have enabled the determination of the precise location of post-translational modification sites, thus providing essential information for understanding the mechanistic details of these regulations. The current challenge is first, to understand the roles of these modifications on the expression and the function of Na(V)1.5, and second, to further identify other chemical modifications. It is postulated that the diversity of phenotypes observed with Na(V)1.5-dependent disorders may partially arise from the complex post-translational modifications of channel protein components.
Collapse
|
22
|
Cerda O, Cáceres M, Park KS, Leiva-Salcedo E, Romero A, Varela D, Trimmer JS, Stutzin A. Casein kinase-mediated phosphorylation of serine 839 is necessary for basolateral localization of the Ca²⁺-activated non-selective cation channel TRPM4. Pflugers Arch 2014; 467:1723-1732. [PMID: 25231975 DOI: 10.1007/s00424-014-1610-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 10/24/2022]
Abstract
Transient receptor potential melastatin-like 4 (TRPM4) is a Ca(2+)-activated non-selective cation channel expressed in a wide range of human tissues. TRPM4 participates in a variety of physiological processes such as T cell activation, myogenic vasoconstriction, and allergic reactions. TRPM4 Ca(2+) sensitivity is enhanced by calmodulin (CaM) and phosphathydilinositol 4, 5-bisphosphate (PI(4,5)P2) binding, as well as, under certain conditions, PKC activation. However, information as to the mechanisms of modulation of this channel remains unknown, including direct identification of phosphorylation sites on TRPM4 and their role in channel features. Here, we use mass-spectrometric-based proteomic approaches (immunoprecipitation and tandem mass spectrometry) to unambiguously identify S839 as a phosphorylation site present on human TRPM4 expressed in a human cell line. Site-directed mutagenesis employing a serine to alanine mutation to eliminate phosphorylation, and a phospho-mimetic aspartate mutation, as well as biochemical and immunocytochemical experiments, revealed a role for S839 phosphorylation in the basolateral expression of TRPM4 channels in epithelial cells. Moreover, we demonstrated that casein kinase 1 (CK1) phosphorylates S839 and is responsible for the basolateral localization of TRPM4.
Collapse
Affiliation(s)
- Oscar Cerda
- Programa de Biología Celular y Molecular, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile.,Centro de Estudios Moleculares de la Célula (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis CA 95616, USA
| | - Mónica Cáceres
- Programa de Biología Celular y Molecular, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile.,Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis CA 95616, USA
| | - Kang-Sik Park
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis CA 95616, USA.,Department of Physiology, Kyung Hee University School of Medicine, Seoul, South Korea 130-701
| | - Elías Leiva-Salcedo
- Centro de Estudios Moleculares de la Célula (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Aníbal Romero
- Programa de Biología Celular y Molecular, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - Diego Varela
- Centro de Estudios Moleculares de la Célula (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis CA 95616, USA.,Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis CA 95616, USA
| | - Andrés Stutzin
- Centro de Estudios Moleculares de la Célula (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Programa de Fisiopatología, Facultad de Medicina, ICBM, Universidad de Chile, Santiago, Chile
| |
Collapse
|
23
|
NMDA receptors and L-type voltage-gated Ca²⁺ channels mediate the expression of bidirectional homeostatic intrinsic plasticity in cultured hippocampal neurons. Neuroscience 2014; 277:610-23. [PMID: 25086314 DOI: 10.1016/j.neuroscience.2014.07.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 07/22/2014] [Indexed: 11/24/2022]
Abstract
Homeostatic plasticity is engaged when neurons need to stabilize their synaptic strength and excitability in response to acute or prolonged destabilizing changes in global activity. Compared to the extensive studies investigating the molecular mechanisms for homeostatic synaptic plasticity, the mechanism underlying homeostatic intrinsic plasticity is largely unknown. Through whole-cell patch-clamp recording in low-density cultures of dissociated hippocampal neurons, we demonstrate here that prolonged activity blockade induced by the sodium channel blocker tetrodotoxin (TTX) leads to increased action potential firing rates. Conversely, prolonged activity enhancement induced by the A-type gamma-aminobutyric acid receptor antagonist bicuculline (BC) results in decreased firing rates. Prolonged activity enhancement also enhanced potassium (K(+)) current through Kv1 channels, suggesting that changes in K(+) current, in part, mediate stabilization of hippocampal neuronal excitability upon prolonged activity elevation. In contrast to the previous reports showing that L-type voltage-gated calcium (Ca(2+)) channels solely mediate homeostatic regulation of excitatory synaptic strength (Ibata et al., 2008; Goold and Nicoll, 2010), inhibition of N-Methyl-d-aspartate (NMDA) receptors alone mimics the elevation in firing frequency driven by prolonged TTX application, while the decrease in firing rates induced by prolonged BC treatment involves the activity of NMDA receptors and L-type voltage-gated Ca(2+) channels. These results collectively provide strong evidence that alterations in Ca(2+) influx through NMDA receptors and L-type voltage-gated Ca(2+) channels mediate homeostatic intrinsic plasticity in hippocampal neurons in response to prolonged activity changes.
Collapse
|
24
|
Moore CL, Nelson PL, Parelkar NK, Rusch NJ, Rhee SW. Protein kinase A-phosphorylated KV1 channels in PSD95 signaling complex contribute to the resting membrane potential and diameter of cerebral arteries. Circ Res 2014; 114:1258-67. [PMID: 24585759 DOI: 10.1161/circresaha.114.303167] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RATIONALE Postsynaptic density-95 (PSD95) is a scaffolding protein that associates with voltage-gated, Shaker-type K(+) (KV1) channels and promotes the expression of KV1 channels in vascular smooth muscle cells of the cerebral (cVSMCs) circulation. However, the physiological role of PSD95 in mediating molecular signaling in cVSMCs is unknown. OBJECTIVE We explored whether a specific interaction between PSD95 and KV1 channels enables protein kinase A phosphorylation of KV1 channels in cVSMCs to promote vasodilation. METHODS AND RESULTS Rat cerebral arteries were used for analyses. A membrane-permeable peptide (KV1-C peptide) corresponding to the postsynaptic density-95, discs large, zonula occludens-1 binding motif in the C terminus of KV1.2α was designed as a dominant-negative peptide to disrupt the association of KV1 channels with PSD95. Application of KV1-C peptide to cannulated, pressurized cerebral arteries rapidly induced vasoconstriction and depolarized cVSMCs. These events corresponded to reduced coimmunoprecipitation of the PSD95 and KV1 proteins without altering surface expression. Middle cerebral arterioles imaged in situ through cranial window also constricted rapidly in response to local application of KV1-C peptide. Patch-clamp recordings confirmed that KV1-C peptide attenuates KV1 channel blocker (5-(4-phenylalkoxypsoralen))-sensitive current in cVSMCs. Western blots using a phospho-protein kinase A substrate antibody revealed that cerebral arteries exposed to KV1-C peptide showed markedly less phosphorylation of KV1.2α subunits. Finally, phosphatase inhibitors blunted both KV1-C peptide-mediated and protein kinase A inhibitor peptide-mediated vasoconstriction. CONCLUSIONS These findings provide initial evidence that protein kinase A phosphorylation of KV1 channels is enabled by a dynamic association with PSD95 in cerebral arteries and suggest that a disruption of such association may compromise cerebral vasodilation and blood flow.
Collapse
Affiliation(s)
- Christopher L Moore
- From the Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock (C.L.M., P.L.N., N.J.R., S.W.R.); and University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City (N.K.P.)
| | | | | | | | | |
Collapse
|
25
|
Bae SH, Kim DH, Shin SK, Choi JS, Park KS. Src regulates membrane trafficking of the Kv3.1b channel. FEBS Lett 2013; 588:86-91. [PMID: 24291260 DOI: 10.1016/j.febslet.2013.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/01/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
Abstract
The Kv3.1 channel plays a crucial role in regulating the high-frequency firing properties of neurons. Here, we determined whether Src regulates the subcellular distributions of the Kv3.1b channel. Co-expression of active Src induced a dramatic redistribution of Kv3.1b to the endoplasmic reticulum. Furthermore, co-expression of the Kv3.1b channel with active Src induced a remarkable decrease in the pool of Kv3.1b at the cell surface. Moreover, the co-expression of active Src results in a significant decrease in the peak current densities of the Kv3.1b channel, and a substantial alteration in the voltage dependence of its steady-state inactivation. Taken together, these results indicate that Src kinase may play an important role in regulating membrane trafficking of Kv3.1b channels.
Collapse
Affiliation(s)
- Seong Han Bae
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
| | - Dong Hyun Kim
- College of Pharmacy, Catholic University of Korea, Bucheon 420-743, Gyeonggi-Do, South Korea
| | - Seok Kyo Shin
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
| | - Jin Sung Choi
- College of Pharmacy, Catholic University of Korea, Bucheon 420-743, Gyeonggi-Do, South Korea
| | - Kang-Sik Park
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea; Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul 130-701, South Korea.
| |
Collapse
|
26
|
Liu Q, Lee E, Davis RL. Heterogeneous intrinsic excitability of murine spiral ganglion neurons is determined by Kv1 and HCN channels. Neuroscience 2013; 257:96-110. [PMID: 24200924 DOI: 10.1016/j.neuroscience.2013.10.065] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 10/02/2013] [Accepted: 10/26/2013] [Indexed: 02/03/2023]
Abstract
The spiral ganglion conveys afferent auditory information predominantly through a single class of type I neurons that receive signals from inner hair cell sensory receptors. These auditory primary afferents, like in other systems (Puopolo and Belluzzi, 1998; Gascon and Moqrich, 2010; Leao et al., 2012) possess a marked diversity in their electrophysiological features (Taberner and Liberman, 2005). Consistent with these observations, when the auditory primary afferents were assessed in neuronal explants separated from their peripheral and central targets it was found that individual neurons were markedly heterogeneous in their endogenous electrophysiological features. One aspect of this heterogeneity, obvious throughout the ganglion, was their wide range of excitability as assessed by voltage threshold measurements (Liu and Davis, 2007). Thus, while neurons in the base differed significantly from apical and middle neurons in their voltage thresholds, each region showed distinctly wide ranges of values. To determine whether the resting membrane potentials (RMPs) of these neurons correlate with the threshold distribution and to identify the ion channel regulatory elements underlying heterogeneous neuronal excitability in the ganglion, patch-clamp recordings were made from postnatal day (P5-8) murine spiral ganglion neurons in vitro. We found that RMP mirrored the tonotopic threshold distribution, and contributed an additional level of heterogeneity in each cochlear location. Pharmacological experiments further indicated that threshold and RMP was coupled through the Kv1 current, which had a dual impact on both electrophysiological parameters. Whereas, hyperpolarization-activated cationic channels decoupled these two processes by primarily affecting RMP without altering threshold level. Thus, beyond mechanical and synaptic specializations, ion channel regulation of intrinsic membrane properties imbues spiral ganglion neurons with different excitability levels, a feature that contributes to primary auditory afferent diversity.
Collapse
Affiliation(s)
- Q Liu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - E Lee
- Rutgers University, New Jersey Medical School, Newark, NJ 07746, USA
| | - R L Davis
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
27
|
Wang W, Kim HJ, Lv P, Tempel B, Yamoah EN. Association of the Kv1 family of K+ channels and their functional blueprint in the properties of auditory neurons as revealed by genetic and functional analyses. J Neurophysiol 2013; 110:1751-64. [PMID: 23864368 DOI: 10.1152/jn.00290.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Developmental plasticity in spiral ganglion neurons (SGNs) ensues from profound alterations in the functional properties of the developing hair cell (HC). For example, prehearing HCs are spontaneously active. However, at the posthearing stage, HC membrane properties transition to graded receptor potentials. The dendrotoxin (DTX)-sensitive Kv1 channel subunits (Kv1.1, 1.2, and 1.6) shape the firing properties and membrane potential of SGNs, and the expression of the channel undergoes developmental changes. Because of the stochastic nature of Kv subunit heteromultimerization, it has been difficult to determine physiologically relevant subunit-specific interactions and their functions in the underlying mechanisms of Kv1 channel plasticity in SGNs. Using Kcna2 null mutant mice, we demonstrate a surprising paradox in changes in the membrane properties of SGNs. The resting membrane potential of Kcna2(-/-) SGNs was significantly hyperpolarized compared with that of age-matched wild-type (WT) SGNs. Analyses of outward currents in the mutant SGNs suggest an apparent approximately twofold increase in outward K(+) currents. We show that in vivo and in vitro heteromultimerization of Kv1.2 and Kv1.4 α-subunits underlies the striking and unexpected alterations in the properties of SGNs. The results suggest that heteromeric interactions of Kv1.2 and Kv1.4 dominate the defining features of Kv1 channels in SGNs.
Collapse
Affiliation(s)
- Wenying Wang
- Program in Communication Science, Center for Neuroscience, University of California, Davis, School of Medicine, Davis, California
| | | | | | | | | |
Collapse
|
28
|
Cerny AC, Oberacker T, Pfannstiel J, Weigold S, Will C, Huber A. Mutation of light-dependent phosphorylation sites of the Drosophila transient receptor potential-like (TRPL) ion channel affects its subcellular localization and stability. J Biol Chem 2013; 288:15600-13. [PMID: 23592784 DOI: 10.1074/jbc.m112.426981] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Drosophila phototransduction cascade terminates in the opening of the ion channel transient receptor potential (TRP) and TRP-like (TRPL). Contrary to TRP, TRPL undergoes light-dependent subcellular trafficking between rhabdomeric photoreceptor membranes and an intracellular storage compartment, resulting in long term light adaptation. Here, we identified in vivo phosphorylation sites of TRPL that affect TRPL stability and localization. Quantitative mass spectrometry revealed a light-dependent change in the TRPL phosphorylation pattern. Mutation of eight C-terminal phosphorylation sites neither affected multimerization of the channels nor the electrophysiological response of flies expressing the mutated channels. However, these mutations resulted in mislocalization and enhanced degradation of TRPL after prolonged dark-adaptation. Mutation of subsets of the eight C-terminal phosphorylation sites also led to a reduction of TRPL content and partial mislocalization in the dark. This suggests that a light-dependent switch in the phosphorylation pattern of the TRPL channel mediates stable expression of TRPL in the rhabdomeres upon prolonged dark-adaptation.
Collapse
Affiliation(s)
- Alexander C Cerny
- Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Vacher H, Trimmer JS. Trafficking mechanisms underlying neuronal voltage-gated ion channel localization at the axon initial segment. Epilepsia 2013; 53 Suppl 9:21-31. [PMID: 23216576 DOI: 10.1111/epi.12032] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Voltage-gated ion channels are diverse and fundamental determinants of neuronal intrinsic excitability. Voltage-gated K(+) (Kv) and Na(+) (Nav) channels play complex yet fundamentally important roles in determining intrinsic excitability. The Kv and Nav channels located at the axon initial segment (AIS) play a unique and especially important role in generating neuronal output in the form of anterograde axonal and backpropagating action potentials. Aberrant intrinsic excitability in individual neurons within networks contributes to synchronous neuronal activity leading to seizures. Mutations in ion channel genes give rise to a variety of seizure-related "channelopathies," and many of the ion channel subunits associated with epilepsy mutations are localized at the AIS, making this a hotspot for epileptogenesis. Here we review the cellular mechanisms that underlie the trafficking of Kv and Nav channels found at the AIS, and how Kv and Nav channel mutations associated with epilepsy can alter these processes.
Collapse
Affiliation(s)
- Helene Vacher
- CRN2M CNRS UMR7286, Aix-Marseille University, Marseille, France
| | | |
Collapse
|
30
|
Schindler J, Ye J, Jensen ON, Nothwang HG. Monitoring the native phosphorylation state of plasma membrane proteins from a single mouse cerebellum. J Neurosci Methods 2012; 213:153-64. [PMID: 23246975 DOI: 10.1016/j.jneumeth.2012.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 10/03/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022]
Abstract
Neuronal processing in the cerebellum involves the phosphorylation and dephosphorylation of various plasma membrane proteins such as AMPA or NMDA receptors. Despite the importance of changes in phosphorylation pattern, no global phospho-proteome analysis has yet been performed. As plasma membrane proteins are major targets of the signalling cascades, we developed a protocol to monitor their phosphorylation state starting from a single mouse cerebellum. An aqueous polymer two-phase system was used to enrich for plasma membrane proteins. Subsequently, calcium phosphate precipitation, immobilized metal affinity chromatography, and TiO(2) were combined to a sequential extraction procedure prior to mass spectrometric analyses. This strategy resulted in the identification of 1501 different native phosphorylation sites in 507 different proteins. 765 (51%) of these phosphorylation sites were localized with a confidence level of 99% or higher. 41.4% of the identified proteins were allocated to the plasma membrane and about half of the phosphorylation sites have not been reported previously. A bioinformatic screen for 12 consensus sequences identified putative kinases for 642 phosphorylation sites. In summary, the protocol deployed here identified several hundred novel phosphorylation sites of cerebellar proteins. Furthermore, it provides a valuable tool to monitor the plasma membrane proteome from any small brain samples of interest under differing physiological or pathophysiological conditions.
Collapse
Affiliation(s)
- Jens Schindler
- Neurogenetics Group, University of Oldenburg, Oldenburg, Germany.
| | | | | | | |
Collapse
|
31
|
Moritz AE, Foster JD, Gorentla BK, Mazei-Robison MS, Yang JW, Sitte HH, Blakely RD, Vaughan RA. Phosphorylation of dopamine transporter serine 7 modulates cocaine analog binding. J Biol Chem 2012; 288:20-32. [PMID: 23161550 DOI: 10.1074/jbc.m112.407874] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
As an approach to elucidating dopamine transporter (DAT) phosphorylation characteristics, we examined in vitro phosphorylation of a recombinant rat DAT N-terminal peptide (NDAT) using purified protein kinases. We found that NDAT becomes phosphorylated at single distinct sites by protein kinase A (Ser-7) and calcium-calmodulin-dependent protein kinase II (Ser-13) and at multiple sites (Ser-4, Ser-7, and Ser-13) by protein kinase C (PKC), implicating these residues as potential sites of DAT phosphorylation by these kinases. Mapping of rat striatal DAT phosphopeptides by two-dimensional thin layer chromatography revealed basal and PKC-stimulated phosphorylation of the same peptide fragments and comigration of PKC-stimulated phosphopeptide fragments with NDAT Ser-7 phosphopeptide markers. We further confirmed by site-directed mutagenesis and mass spectrometry that Ser-7 is a site for PKC-stimulated phosphorylation in heterologously expressed rat and human DATs. Mutation of Ser-7 and nearby residues strongly reduced the affinity of rat DAT for the cocaine analog (-)-2β-carbomethoxy-3β-(4-fluorophenyl) tropane (CFT), whereas in rat striatal tissue, conditions that promote DAT phosphorylation caused increased CFT affinity. Ser-7 mutation also affected zinc modulation of CFT binding, with Ala and Asp substitutions inducing opposing effects. These results identify Ser-7 as a major site for basal and PKC-stimulated phosphorylation of native and expressed DAT and suggest that Ser-7 phosphorylation modulates transporter conformational equilibria, shifting the transporter between high and low affinity cocaine binding states.
Collapse
Affiliation(s)
- Amy E Moritz
- Department of Biochemistry and Molecular Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203-9037, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Marionneau C, Lichti CF, Lindenbaum P, Charpentier F, Nerbonne JM, Townsend RR, Mérot J. Mass spectrometry-based identification of native cardiac Nav1.5 channel α subunit phosphorylation sites. J Proteome Res 2012; 11:5994-6007. [PMID: 23092124 DOI: 10.1021/pr300702c] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cardiac voltage-gated Na+ (Nav) channels are key determinants of action potential waveforms, refractoriness and propagation, and Nav1.5 is the main Nav pore-forming (α) subunit in the mammalian heart. Although direct phosphorylation of the Nav1.5 protein has been suggested to modulate various aspects of Nav channel physiology and pathophysiology, native Nav1.5 phosphorylation sites have not been identified. In the experiments here, a mass spectrometry (MS)-based proteomic approach was developed to identify native Nav1.5 phosphorylation sites directly. Using an anti-NavPAN antibody, Nav channel complexes were immunoprecipitated from adult mouse cardiac ventricles. The MS analyses revealed that this antibody immunoprecipitates several Nav α subunits in addition to Nav1.5, as well as several previously identified Nav channel associated/regulatory proteins. Label-free comparative and data-driven phosphoproteomic analyses of purified cardiac Nav1.5 protein identified 11 phosphorylation sites, 8 of which are novel. All the phosphorylation sites identified except one in the N-terminus are in the first intracellular linker loop, suggesting critical roles for this region in phosphorylation-dependent cardiac Nav channel regulation. Interestingly, commonly used prediction algorithms did not reliably predict these newly identified in situ phosphorylation sites. Taken together, the results presented provide the first in situ map of basal phosphorylation sites on the mouse cardiac Nav1.5 α subunit.
Collapse
|
33
|
Foster JD, Yang JW, Moritz AE, Challasivakanaka S, Smith MA, Holy M, Wilebski K, Sitte HH, Vaughan RA. Dopamine transporter phosphorylation site threonine 53 regulates substrate reuptake and amphetamine-stimulated efflux. J Biol Chem 2012; 287:29702-12. [PMID: 22722938 PMCID: PMC3436161 DOI: 10.1074/jbc.m112.367706] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In the central nervous system, levels of extraneuronal dopamine are controlled primarily by the action of the dopamine transporter (DAT). Multiple signaling pathways regulate transport activity, substrate efflux, and other DAT functions through currently unknown mechanisms. DAT is phosphorylated by protein kinase C within a serine cluster at the distal end of the cytoplasmic N terminus, whereas recent work in model cells revealed proline-directed phosphorylation of rat DAT at membrane-proximal residue Thr(53). In this report, we use mass spectrometry and a newly developed phospho-specific antibody to positively identify DAT phosphorylation at Thr(53) in rodent striatal tissue and heterologous expression systems. Basal phosphorylation of Thr(53) occurred with a stoichiometry of ~50% and was strongly increased by phorbol esters and protein phosphatase inhibitors, demonstrating modulation of the site by signaling pathways that impact DAT activity. Mutations of Thr(53) to prevent phosphorylation led to reduced dopamine transport V(max) and total apparent loss of amphetamine-stimulated substrate efflux, supporting a major role for this residue in the transport kinetic mechanism.
Collapse
Affiliation(s)
- James D Foster
- Department of Biochemistry and Molecular Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202-9037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Manning CF, Bundros AM, Trimmer JS. Benefits and pitfalls of secondary antibodies: why choosing the right secondary is of primary importance. PLoS One 2012; 7:e38313. [PMID: 22675541 PMCID: PMC3365890 DOI: 10.1371/journal.pone.0038313] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/08/2012] [Indexed: 01/21/2023] Open
Abstract
Simultaneous labeling of multiple targets in a single sample, or multiplexing, is a powerful approach to directly compare the amount, localization and/or molecular properties of different targets in the same sample. Here we highlight the robust reliability of the simultaneous use of multiple mouse monoclonal antibodies (mAbs) of different immunoglobulin G (IgG) subclasses in a wide variety of multiplexing applications employing anti-mouse IgG subclass-specific secondary antibodies (2°Abs). We also describe the unexpected finding that IgG subclass-specific 2°Abs are superior to general anti-mouse IgG 2°Abs in every tested application in which mouse mAbs were used. This was due to a detection bias of general anti-mouse IgG-specific 2°Abs against mAbs of the most common mouse IgG subclass, IgG1, and to a lesser extent IgG2b mAbs. Thus, when using any of numerous mouse mAbs available through commercial and non-profit sources, for cleaner and more robust results each mAb should be detected with its respective IgG subclass-specific 2°Ab and not a general anti-mouse IgG-specific 2°Ab.
Collapse
Affiliation(s)
- Colleen F. Manning
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, United States of America
| | - Angeliki M. Bundros
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, United States of America
| | - James S. Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, United States of America
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
35
|
Song MY, Hong C, Bae SH, So I, Park KS. Dynamic modulation of the kv2.1 channel by SRC-dependent tyrosine phosphorylation. J Proteome Res 2012; 11:1018-26. [PMID: 22106938 PMCID: PMC3272096 DOI: 10.1021/pr200770v] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The voltage-gated K(+) channel Kv2.1 is expressed as a highly phosphorylated protein in most central neurons, where it plays a key role in regulating neuronal membrane excitability. Previous studies have shown that Kv2.1 channel activity is upregulated by Src-mediated phosphorylation through an unknown mechanism. However, a systematic analysis of the molecular mechanism of Kv2.1 channel phosphorylation by Src is lacking. Here, we show that tyrosine phosphorylation by Src plays a fundamental role in regulating Kv2.1-mediated K(+) current enhancement. We found that the level of expression of the Kv2.1 protein is increased by Src kinase. Using mass spectrometric proteomic techniques, we identified two novel phosphotyrosine sites, Y686 and Y810, in the cytoplasmic domains of Kv2.1. We found that Src-dependent phosphorylation at these sites affects Kv2.1 through distinct regulatory mechanisms. Whereas phosphorylation at Y686 regulates Kv2.1 activity similarly to the known site Y124, phosphorylation at Y810 plays a significant role in regulating the intracellular trafficking of Kv2.1 channels. Our results show that these two novel tyrosine phosphorylation sites of Kv2.1 are crucial to regulating diverse aspects of Kv2.1 channel function and provide novel insights into molecular mechanisms for the regulation of Src-dependent modulation of Kv2.1 channels.
Collapse
Affiliation(s)
- Min-Young Song
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
- Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
| | - Chansik Hong
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Seong Han Bae
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
- Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Kang-Sik Park
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
- Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul 130-701, South Korea
| |
Collapse
|
36
|
Nelson C, Glitsch MD. Lack of kinase regulation of canonical transient receptor potential 3 (TRPC3) channel-dependent currents in cerebellar Purkinje cells. J Biol Chem 2011; 287:6326-35. [PMID: 22207762 DOI: 10.1074/jbc.m111.246553] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Canonical transient receptor potential (TRPC) channels are widely expressed in the brain and play several roles in development and normal neuronal function. In the cerebellum, Purkinje cell TRPC3 channels underlie the slow excitatory postsynaptic potential observed after parallel fiber stimulation. In these cells TRPC3 channel opening requires stimulation of metabotropic glutamate receptor 1, activation of which can also lead to the induction of long term depression (LTD), which underlies cerebellar motor learning. LTD induction requires protein kinase C (PKC) and protein kinase G (PKG) activation, and although PKC phosphorylation targets are well established, virtually nothing is known about PKG targets in LTD. Because TRPC3 channels are inhibited after phosphorylation by PKC and PKG in expression systems, we examined whether native TRPC3 channels in Purkinje cells are a target for PKG or PKC, thereby contributing to cerebellar LTD. We find that in Purkinje cells, activation of TRPC3-dependent currents is not inhibited by conventional PKC or PKG to any significant extent and that inhibition of these kinases does not significantly impact on TRPC3-mediated currents either. Based on these and previous findings, we propose that TRPC3-dependent currents may differ significantly in their regulation from those overexpressed in expression systems.
Collapse
Affiliation(s)
- Charmaine Nelson
- Department of Physiology, Anatomy, and Genetics, Sherrington Building, Parks Road, Oxford Univerity, Oxford OX1 3PT, United Kingdom
| | | |
Collapse
|
37
|
Liao L, Sando RC, Farnum JB, Vanderklish PW, Maximov A, Yates JR. 15N-labeled brain enables quantification of proteome and phosphoproteome in cultured primary neurons. J Proteome Res 2011; 11:1341-53. [PMID: 22070516 DOI: 10.1021/pr200987h] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Terminally differentiated primary cells represent a valuable in vitro model to study signaling events associated within a specific tissue. Quantitative proteomic methods using metabolic labeling in primary cells encounter labeling efficiency issues hindering the use of these cells. Here we developed a method to quantify the proteome and phosphoproteome of cultured neurons using (15)N-labeled brain tissue as an internal standard and applied this method to determine how an inhibitor of an excitatory neural transmitter receptor, phencyclidine (PCP), affects the global phosphoproteome of cortical neurons. We identified over 10,000 phosphopeptides and made accurate quantitative measurements of the neuronal phosphoproteome after neuronal inhibition. We show that short PCP treatments lead to changes in phosphorylation for 7% of neuronal phosphopeptides and that prolonged PCP treatment alters the total levels of several proteins essential for synaptic transmission and plasticity and leads to a massive reduction in the synaptic strength of inhibitory synapses. The results provide valuable insights into the dynamics of molecular networks implicated in PCP-mediated NMDA receptor inhibition and sensorimotor deficits.
Collapse
Affiliation(s)
- Lujian Liao
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, United States
| | | | | | | | | | | |
Collapse
|
38
|
Diverse roles for auxiliary subunits in phosphorylation-dependent regulation of mammalian brain voltage-gated potassium channels. Pflugers Arch 2011; 462:631-43. [PMID: 21822597 DOI: 10.1007/s00424-011-1004-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 07/22/2011] [Accepted: 07/22/2011] [Indexed: 10/17/2022]
Abstract
Voltage-gated ion channels are a diverse family of signaling proteins that mediate rapid electrical signaling events. Among these, voltage-gated potassium or Kv channels are the most diverse partly due to the large number of principal (or α) subunits and auxiliary subunits that can assemble in different combinations to generate Kv channel complexes with distinct structures and functions. The diversity of Kv channels underlies much of the variability in the active properties between different mammalian central neurons and the dynamic changes that lead to experience-dependent plasticity in intrinsic excitability. Recent studies have revealed that Kv channel α subunits and auxiliary subunits are extensively phosphorylated, contributing to additional structural and functional diversity. Here, we highlight recent studies that show that auxiliary subunits exert some of their profound effects on dendritic Kv4 and axonal Kv1 channels through phosphorylation-dependent mechanisms, either due to phosphorylation on the auxiliary subunit itself or by influencing the extent and/or impact of α subunit phosphorylation. The complex effects of auxiliary subunits and phosphorylation provide a potent mechanism to generate additional diversity in the structure and function of Kv4 and Kv1 channels, as well as allowing for dynamic reversible regulation of these important ion channels.
Collapse
|
39
|
Jensen CS, Rasmussen HB, Misonou H. Neuronal trafficking of voltage-gated potassium channels. Mol Cell Neurosci 2011; 48:288-97. [PMID: 21627990 DOI: 10.1016/j.mcn.2011.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 05/01/2011] [Accepted: 05/16/2011] [Indexed: 11/28/2022] Open
Abstract
The computational ability of CNS neurons depends critically on the specific localization of ion channels in the somatodendritic and axonal membranes. Neuronal dendrites receive synaptic inputs at numerous spines and integrate them in time and space. The integration of synaptic potentials is regulated by voltage-gated potassium (Kv) channels, such as Kv4.2, which are specifically localized in the dendritic membrane. The synaptic potentials eventually depolarize the membrane of the axon initial segment, thereby activating voltage-gated sodium channels to generate action potentials. Specific Kv channels localized in the axon initial segment, such as Kv1 and Kv7 channels, determine the shape and the rate of action potentials. Kv1 and Kv7 channels present at or near nodes of Ranvier and in presynaptic terminals also influence the propagation of action potentials and neurotransmitter release. The physiological significance of proper Kv channel localization is emphasized by the fact that defects in the trafficking of Kv channels are observed in several neurological disorders including epilepsy. In this review, we will summarize the current understanding of the mechanisms of Kv channel trafficking and discuss how they contribute to the establishment and maintenance of the specific localization of Kv channels in neurons.
Collapse
Affiliation(s)
- Camilla S Jensen
- Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
40
|
Cerda O, Baek JH, Trimmer JS. Mining recent brain proteomic databases for ion channel phosphosite nuggets. ACTA ACUST UNITED AC 2010; 137:3-16. [PMID: 21149544 PMCID: PMC3010060 DOI: 10.1085/jgp.201010555] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Voltage-gated ion channels underlie electrical activity of neurons and are dynamically regulated by diverse cell signaling pathways that alter their phosphorylation state. Recent global mass spectrometric–based analyses of the mouse brain phosphoproteome have yielded a treasure trove of new data as to the extent and nature of phosphorylation of numerous ion channel principal or α subunits in mammalian brain. Here we compile and review data on 347 phosphorylation sites (261 unique) on 42 different voltage-gated ion channel α subunits that were identified in these recent studies. Researchers in the ion channel field can now begin to explore the role of these novel in vivo phosphorylation sites in the dynamic regulation of the localization, activity, and expression of brain ion channels through multisite phosphorylation of their principal subunits.
Collapse
Affiliation(s)
- Oscar Cerda
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA
| | | | | |
Collapse
|
41
|
Role of the ubiquitin system in regulating ion transport. Pflugers Arch 2010; 461:1-21. [PMID: 20972579 DOI: 10.1007/s00424-010-0893-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 10/04/2010] [Accepted: 10/04/2010] [Indexed: 12/22/2022]
Abstract
Ion channels and transporters play a critical role in ion and fluid homeostasis and thus in normal animal physiology and pathology. Tight regulation of these transmembrane proteins is therefore essential. In recent years, many studies have focused their attention on the role of the ubiquitin system in regulating ion channels and transporters, initialed by the discoveries of the role of this system in processing of Cystic Fibrosis Transmembrane Regulator (CFTR), and in regulating endocytosis of the epithelial Na(+) channel (ENaC) by the Nedd4 family of ubiquitin ligases (mainly Nedd4-2). In this review, we discuss the role of the ubiquitin system in ER Associated Degradation (ERAD) of ion channels, and in the regulation of endocytosis and lysosomal sorting of ion channels and transporters, focusing primarily in mammalian cells. We also briefly discuss the role of ubiquitin like molecules (such as SUMO) in such regulation, for which much less is known so far.
Collapse
|
42
|
Marionneau C, Townsend RR, Nerbonne JM. Proteomic analysis highlights the molecular complexities of native Kv4 channel macromolecular complexes. Semin Cell Dev Biol 2010; 22:145-52. [PMID: 20959143 DOI: 10.1016/j.semcdb.2010.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Revised: 10/07/2010] [Accepted: 10/08/2010] [Indexed: 01/24/2023]
Abstract
Voltage-gated K(+) (Kv) channels are key determinants of membrane excitability in the nervous and cardiovascular systems, functioning to control resting membrane potentials, shape action potential waveforms and influence the responses to neurotransmitters and neurohormones. Consistent with this functional diversity, multiple types of Kv currents, with distinct biophysical properties and cellular/subcellular distributions, have been identified. Rapidly activating and inactivating Kv currents, typically referred to as I(A) (A-type) in neurons, for example, regulate repetitive firing rates, action potential back-propagation (into dendrites) and modulate synaptic responses. Currents with similar properties, referred to as I(to,f) (fast transient outward), expressed in cardiomyocytes, control the early phase of myocardial action potential repolarization. A number of studies have demonstrated critical roles for pore-forming (α) subunits of the Kv4 subfamily in the generation of native neuronal I(A) and cardiac I(to,f) channels. Studies in heterologous cells have also suggested important roles for a number of Kv channel accessory and regulatory proteins in the generation of functional I(A) and I(to,f) channels. Quantitative mass spectrometry-based proteomic analysis is increasingly recognized as a rapid and, importantly, unbiased, approach to identify the components of native macromolecular protein complexes. The recent application of proteomic approaches to identify the components of native neuronal (and cardiac) Kv4 channel complexes has revealed even greater complexity than anticipated. The continued emphasis on development of improved biochemical and analytical proteomic methods seems certain to accelerate progress and to provide important new insights into the molecular determinants of native ion channel protein complexes.
Collapse
Affiliation(s)
- Céline Marionneau
- L'Institut du Thorax, INSERM UMR-915, Institut de Recherche Thérapeutique-Université de Nantes, Nantes, France
| | | | | |
Collapse
|
43
|
Baek JH, Cerda O, Trimmer JS. Mass spectrometry-based phosphoproteomics reveals multisite phosphorylation on mammalian brain voltage-gated sodium and potassium channels. Semin Cell Dev Biol 2010; 22:153-9. [PMID: 20932926 DOI: 10.1016/j.semcdb.2010.09.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 09/23/2010] [Accepted: 09/28/2010] [Indexed: 12/19/2022]
Abstract
Voltage-gated sodium and potassium channels underlie electrical activity of neurons, and are dynamically regulated by diverse cell signaling pathways that ultimately exert their effects by altering the phosphorylation state of channel subunits. Recent mass spectrometric-based studies have led to a new appreciation of the extent and nature of phosphorylation of these ion channels in mammalian brain. This has allowed for new insights into how neurons dynamically regulate the localization, activity and expression through multisite ion channel phosphorylation.
Collapse
Affiliation(s)
- Je-Hyun Baek
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616-8519, United States
| | | | | |
Collapse
|
44
|
Mierzwa AJ, Arevalo JC, Schiff R, Chao MV, Rosenbluth J. Role of transverse bands in maintaining paranodal structure and axolemmal domain organization in myelinated nerve fibers: effect on longevity in dysmyelinated mutant mice. J Comp Neurol 2010; 518:2841-53. [PMID: 20506478 DOI: 10.1002/cne.22367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The consequences of dysmyelination are poorly understood and vary widely in severity. The shaking mouse, a quaking allele, is characterized by severe central nervous system (CNS) dysmyelination and demyelination, a conspicuous action tremor, and seizures in approximately 25% of animals, but with normal muscle strength and a normal lifespan. In this study we compare this mutant with other dysmyelinated mutants including the ceramide sulfotransferase deficient (CST-/-) mouse, which are more severely affected behaviorally, to determine what might underlie the differences between them with respect to behavior and longevity. Examination of the paranodal junctional region of CNS myelinated fibers shows that "transverse bands," a component of the junction, are present in nearly all shaking paranodes but in only a minority of CST-/- paranodes. The number of terminal loops that have transverse bands within a paranode and the number of transverse bands per unit length are only moderately reduced in the shaking mutant, compared with controls, but markedly reduced in CST-/- mice. Immunofluorescence studies also show that although the nodes of the shaking mutant are somewhat longer than normal, Na(+) and K(+) channels remain separated, distinguishing this mutant from CST-/- mice and others that lack transverse bands. We conclude that the essential difference between the shaking mutant and others more severely affected is the presence of transverse bands, which serve to stabilize paranodal structure over time as well as the organization of the axolemmal domains, and that differences in the prevalence of transverse bands underlie the marked differences in progressive neurological impairment and longevity among dysmyelinated mouse mutants.
Collapse
Affiliation(s)
- Amanda J Mierzwa
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
45
|
Yao H, Zhou K, Yan D, Li M, Wang Y. The Kv2.1 channels mediate neuronal apoptosis induced by excitotoxicity. J Neurochem 2010. [DOI: 10.1111/j.0022-3042.2008.05834.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Berendt FJ, Park KS, Trimmer JS. Multisite phosphorylation of voltage-gated sodium channel alpha subunits from rat brain. J Proteome Res 2010; 9:1976-84. [PMID: 20131913 DOI: 10.1021/pr901171q] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reversible phosphorylation of ion channels underlies cellular plasticity in mammalian neurons. Voltage-gated sodium or Nav channels underlie action potential initiation and propagation, dendritic excitability, and many other aspects of neuronal excitability. Various protein kinases have been suggested to phosphorylate the primary or alpha subunit of Nav channels, affecting diverse aspects of channel function. Previous studies of Nav alpha subunit phosphorylation have led to the identification of a small set of phosphorylation sites important in mediating diverse aspects of Nav channel function. Here we use nanoflow liquid chromatography tandem mass spectrometry (nano-LC MS/MS) on Nav alpha subunits affinity-purified from rat brain with two distinct monoclonal antibodies to identify 15 phosphorylation sites on Nav1.2, 12 of which have not been previously reported. We also found 3 novel phosphorylation sites on Nav1.1. In general, commonly used phosphorylation site prediction algorithms did not accurately predict these novel in vivo phosphorylation sites. Our results demonstrate that specific Nav alpha subunits isolated from rat brain are highly phosphorylated, and suggest extensive modulation of Nav channel activity in mammalian brain. Identification of phosphorylation sites using monoclonal antibody-based immunopurification and mass spectrometry is an effective approach to define the phosphorylation status of Nav channels and other important membrane proteins in mammalian brain.
Collapse
Affiliation(s)
- Frank J Berendt
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, School of Medicine, University of California, Davis, California 95616-8519, USA
| | | | | |
Collapse
|
47
|
Cerda O, Trimmer JS. Analysis and functional implications of phosphorylation of neuronal voltage-gated potassium channels. Neurosci Lett 2010; 486:60-7. [PMID: 20600597 DOI: 10.1016/j.neulet.2010.06.064] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 06/16/2010] [Accepted: 06/22/2010] [Indexed: 11/30/2022]
Abstract
Phosphorylation is the most common and abundant post-translational modification to eukaryotic proteins, regulating a plethora of dynamic cellular processes. Here, we review and discuss recent advances in our knowledge of the breadth and importance of reversible phosphorylation in regulating the expression, localization and function of mammalian neuronal voltage-gated potassium (Kv) channels, key regulators of neuronal function. We highlight the role of modern mass spectrometric techniques and phosphospecific antibodies in revealing the extent and nature of phosphorylation at specific sites in Kv channels. We also emphasize the role of reversible phosphorylation in dynamically regulating diverse aspects of Kv channel biology. Finally, we discuss as important future directions the determination of the mechanistic basis for how altering phosphorylation state affects Kv channel expression, localization and function, the nature of macromolecular signaling complexes containing Kv channels and enzymes regulating their phosphorylation state, and the specific role of Kv channel phosphorylation in regulating neuronal function during physiological and pathophysiological events.
Collapse
Affiliation(s)
- Oscar Cerda
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616-8519, United States
| | | |
Collapse
|
48
|
Voolstra O, Beck K, Oberegelsbacher C, Pfannstiel J, Huber A. Light-dependent phosphorylation of the drosophila transient receptor potential ion channel. J Biol Chem 2010; 285:14275-84. [PMID: 20215118 PMCID: PMC2863191 DOI: 10.1074/jbc.m110.102053] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 03/04/2010] [Indexed: 11/06/2022] Open
Abstract
The Drosophila phototransduction cascade terminates in the opening of an ion channel, designated transient receptor potential (TRP). TRP has been shown to become phosphorylated in vitro, suggesting regulation of the ion channel through posttranslational modification. However, except for one phosphorylation site, Ser(982), which was analyzed by functional in vivo studies (Popescu, D. C., Ham, A. J., and Shieh, B. H. (2006) J. Neurosci. 26, 8570-8577), nothing is known about the role of TRP phosphorylation in vivo. Here, we report the identification of 21 TRP phosphorylation sites by a mass spectrometry approach. 20 phosphorylation sites are located in the C-terminal portion of the channel, and one site is located near the N terminus. All 21 phosphorylation sites were also identified in the inaC(P209) mutant, indicating that phosphorylation of TRP at these sites occurred independently from the eye-enriched protein kinase C. Relative quantification of phosphopeptides revealed that at least seven phosphorylation sites were predominantly phosphorylated in the light, whereas one site, Ser(936), was predominantly phosphorylated in the dark. We show that TRP phosphorylated at Ser(936) was located in the rhabomere. Light-dependent changes in the phosphorylation state of this site occurred within minutes. The dephosphorylation of TRP at Ser(936) required activation of the phototransduction cascade.
Collapse
Affiliation(s)
- Olaf Voolstra
- Department of Biosensorics, Institute of Physiology, Germany.
| | | | | | | | | |
Collapse
|
49
|
Regulatory role of the extreme C-terminal end of the S6 inner helix in C-terminal-truncated Kv1.2 channel activation. Cell Biol Int 2010; 34:433-9. [PMID: 19947938 DOI: 10.1042/cbi20090009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The transmembrane part of the S6 inner helix of the Kv1.2 potassium channel is a pivotal part in sustaining channel activity. However, the role of its extreme C-terminal end, which is located on the cytoplasmic side of the channel, is largely unknown. Here, we investigated the role of the extreme C-terminal end of the S6 inner helix (the HRET region) by constructing a series of C-terminal-truncated mutations related to this region in the C-terminal-truncated Kv1.2 channel. Mutations on Thr421 or Glu420 significantly altered the activation of the truncated channel. Mutations on Arg419 demonstrated that neutral or basic, but not acidic amino acid, is essential at the position for the truncated channel activation, and no functional channel was observed when the channel was truncated from His418. Hence, our results indicate that the extreme C-terminal end of the S6 inner helix plays an important regulatory role in the activation of the C-terminal-truncated Kv1.2 channel.
Collapse
|
50
|
Venugopal A, Chaerkady R, Pandey A. Application of mass spectrometry-based proteomics for biomarker discovery in neurological disorders. Ann Indian Acad Neurol 2010; 12:3-11. [PMID: 20151002 PMCID: PMC2811975 DOI: 10.4103/0972-2327.48845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 10/10/2008] [Accepted: 11/10/2008] [Indexed: 12/03/2022] Open
Abstract
Mass spectrometry-based quantitative proteomics has emerged as a powerful approach that has the potential to accelerate biomarker discovery, both for diagnostic as well as therapeutic purposes. Proteomics has traditionally been synonymous with 2D gels but is increasingly shifting to the use of gel-free systems and liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Quantitative proteomic approaches have already been applied to investigate various neurological disorders, especially in the context of identifying biomarkers from cerebrospinal fluid and serum. This review highlights the scope of different applications of quantitative proteomics in understanding neurological disorders with special emphasis on biomarker discovery.
Collapse
|