51
|
Nakamura TY, Coetzee WA. Functional and pharmacological characterization of a Shal-related K+ channel subunit in Zebrafish. BMC PHYSIOLOGY 2008; 8:2. [PMID: 18261223 PMCID: PMC2270284 DOI: 10.1186/1472-6793-8-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Accepted: 02/08/2008] [Indexed: 12/04/2022]
Abstract
Background K+ channels are diverse; both in terms of their function and their molecular composition. Shal subunits were first described in Drosophila. There are three mammalian orthologs, which are members of the Kv4 subfamily. They are involved in neuronal firing patterns as well as control of the cardiac action potential duration. Results Here, we report the biophysical and pharmacological characterization of zShal3, which is the ortholog of the mammalian Kv4.3 subunit, which in mammals is involved in action potential repolarization and gives rise to neuronal A-type K+ currents involved in somatodendretic signal integration. Conclusion We demonstrate that zShal has similar functional and pharmacological characteristics compared to Kv4.3 and it is similarly regulated by pharmacological agents and by the Kv4 accessory subunit, NCS-1.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Molecular Physiology, National Cardiovascular Center Research Institute, Osaka, Japan.
| | | |
Collapse
|
52
|
Bett GCL, Rasmusson RL. Modification of K+ channel-drug interactions by ancillary subunits. J Physiol 2007; 586:929-50. [PMID: 18096604 DOI: 10.1113/jphysiol.2007.139279] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Reconciling ion channel alpha-subunit expression with native ionic currents and their pharmacological sensitivity in target organs has proved difficult. In native tissue, many K(+) channel alpha-subunits co-assemble with ancillary subunits, which can profoundly affect physiological parameters including gating kinetics and pharmacological interactions. In this review, we examine the link between voltage-gated potassium ion channel pharmacology and the biophysics of ancillary subunits. We propose that ancillary subunits can modify the interaction between pore blockers and ion channels by three distinct mechanisms: changes in (1) binding site accessibility; (2) orientation of pore-lining residues; (3) the ability of the channel to undergo post-binding conformational changes. Each of these subunit-induced changes has implications for gating, drug affinity and use dependence of their respective channel complexes. A single subunit may modulate its associated alpha-subunit by more than one of these mechanisms. Voltage-gated potassium channels are the site of action of many therapeutic drugs. In addition, potassium channels interact with drugs whose primary target is another channel, e.g. the calcium channel blocker nifedipine, the sodium channel blocker quinidine, etc. Even when K(+) channel block is the intended mode of action, block of related channels in non-target organs, e.g. the heart, can result in major and potentially lethal side-effects. Understanding factors that determine specificity, use dependence and other properties of K(+) channel drug binding are therefore of vital clinical importance. Ancillary subunits play a key role in determining these properties in native tissue, and so understanding channel-subunit interactions is vital to understanding clinical pharmacology.
Collapse
Affiliation(s)
- Glenna C L Bett
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, 124 Sherman Hall, State University of New York at Buffalo, Buffalo, NY 14214-3005, USA
| | | |
Collapse
|
53
|
Abstract
In neurons, intracellular calcium signals have crucial roles in activating neurotransmitter release and in triggering alterations in neuronal function. Calmodulin has been widely studied as a Ca(2+) sensor that has several defined roles in neuronal Ca(2+) signalling, but members of the neuronal calcium sensor protein family have also begun to emerge as key components in a number of regulatory pathways and have increased the diversity of neuronal Ca(2+) signalling pathways. The differing properties of these proteins allow them to have discrete, non-redundant functions.
Collapse
Affiliation(s)
- Robert D Burgoyne
- The Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, UK.
| |
Collapse
|
54
|
Pioletti M, Findeisen F, Hura GL, Minor DL. Three-dimensional structure of the KChIP1-Kv4.3 T1 complex reveals a cross-shaped octamer. Nat Struct Mol Biol 2006; 13:987-95. [PMID: 17057713 PMCID: PMC3018330 DOI: 10.1038/nsmb1164] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 10/03/2006] [Indexed: 11/09/2022]
Abstract
Brain I(A) and cardiac I(to) currents arise from complexes containing Kv4 voltage-gated potassium channels and cytoplasmic calcium-sensor proteins (KChIPs). Here, we present X-ray crystallographic and small-angle X-ray scattering data that show that the KChIP1-Kv4.3 N-terminal cytoplasmic domain complex is a cross-shaped octamer bearing two principal interaction sites. Site 1 comprises interactions between a unique Kv4 channel N-terminal hydrophobic segment and a hydrophobic pocket formed by displacement of the KChIP H10 helix. Site 2 comprises interactions between a T1 assembly domain loop and the KChIP H2 helix. Functional and biochemical studies indicate that site 1 influences channel trafficking, whereas site 2 affects channel gating, and that calcium binding is intimately linked to KChIP folding and complex formation. Together, the data resolve how Kv4 channels and KChIPs interact and provide a framework for understanding how KChIPs modulate Kv4 function.
Collapse
Affiliation(s)
- Marta Pioletti
- Cardiovascular Research Institute, Department of Biochemistry, California Institute for Quantitative Biomedical Research, University of California, San Francisco, California 94143-2532, USA
| | | | | | | |
Collapse
|
55
|
Ohya S. [Molecular pharmacological studies on potassium channels and their regulatory molecules]. YAKUGAKU ZASSHI 2006; 126:945-53. [PMID: 17016023 DOI: 10.1248/yakushi.126.945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
K+ channels play important roles in the control of a large variety of physiological functions such as muscle contraction, neurotransmitter release, hormone secretion, and cell proliferation. Over 100 cloned K+ channel pore-forming alpha and accessory beta subunits have been identified so far. Here, we introduce a series of molecular pharmacological and physiological studies on some types of voltage-dependent K+ channels and Ca2+-activated K+ channels. We examined molecular cloning and functional characterization of novel, fast-inactivating, A-type K+ channel alpha (Kv4.3L) and beta (KChIP2S) subunits predominantly expressed in mammalian heart and found the sites in Kv4 channels for 1) the regulation of voltage dependency and 2) the CaMKII phosphorylation in the C-terminal cytoplasmic domain. Moreover, we found that delayed rectifier-type K+ channels (ERG1 and KCNQ) contribute to the resting membrane conductance in vascular and gastrointestinal smooth muscles. The large-conductance Ca2+-activated K+ (BK) channel is ubiquitously expressed and contributes to diverse physiological processes. Recent reports have shown that a BK-like channel (mitoKCa) is expressed in cardiac mitochondria, suggesting that BK channel openers protect mammalian hearts against ischemic injury. Our studies revealed that BKbeta1 interacts with cytochrome c oxidase I (Cco1) in cardiac mitochondria, and that the activation of BK channels by 17beta-estradiol results in a significant increase in the survival rate of ventricular myocytes. These findings suggest that BKbeta1 may play an important role in the regulation of cell respiration in cardiac myocytes and be a target for the modulation by female gonadal hormones.
Collapse
Affiliation(s)
- Susumu Ohya
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Japan.
| |
Collapse
|
56
|
Flaim SN, Giles WR, McCulloch AD. Contributions of sustained INa and IKv43 to transmural heterogeneity of early repolarization and arrhythmogenesis in canine left ventricular myocytes. Am J Physiol Heart Circ Physiol 2006; 291:H2617-29. [PMID: 16829642 DOI: 10.1152/ajpheart.00350.2006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The roles of sustained components of I(Na) and I(Kv43) in shaping the action potentials (AP) of myocytes isolated from the canine left ventricle (LV) have not been studied in detail. Here we investigate the hypothesis that these two currents can contribute substantially to heterogeneity of early repolarization and arrhythmic risk. Quantitative data from voltage-clamp and expression profiling experiments were used to complete meaningful modifications to an existing "local control" model of canine midmyocardial myocyte excitation-contraction coupling for epicardial and endocardial cells. We include 1) heterogeneous I(Kv43), I(Ks), and I(SERCA) density; 2) modulation of I(Kv43) by Kv channel interacting protein type 2 (KChIP2) channel subunits; 3) a possible Ca(2+)-dependent open-state inactivation of I(Kv43); and 4) a sustained component of the inward Na(+) current, I(NaL). The resulting simulations illustrate ways in which KChIP2- and Ca(2+)-dependent control of I(Kv43) can result in a sustained outward current that can neutralize I(NaL) in a rate- and myocyte subtype-dependent manner. Both these currents appear to play significant roles in modulating AP duration and rate dependence in midmyocardial myocytes. Furthermore, an increased ratio of I(Kv43) to I(NaL) is capable of protecting epicardial myocytes from the early afterdepolarizations resulting from the SCN5A-I1768V mutation-induced increase in I(NaL). Experimentally observed transmural differences in Ca(2+) handling, including greater sarcoplasmic reticulum Ca(2+) content and faster Ca(2+) transient decay rates on the epicardium, were recapitulated in our simulations. By design, these models allow upward integration into organ models or may be used as a basis for further investigations into cellular heterogeneities.
Collapse
Affiliation(s)
- Sarah N Flaim
- University of California, San Diego, La Jolla, CA 92093-0412, USA
| | | | | |
Collapse
|
57
|
Fik-Rymarkiewicz E, Duda T, Sharma RK. Novel frequenin-modulated Ca2+-signaling membrane guanylate cyclase (ROS-GC) transduction pathway in bovine hippocampus. Mol Cell Biochem 2006; 291:187-204. [PMID: 16733800 DOI: 10.1007/s11010-006-9215-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Frequenin is a member of the neuronal Ca(2+) sensor protein family, implicated in being the modulator of the neurotransmitter release, potassium channels, phosphatidylinositol signaling pathway and the Ca(2+)-dependent exocytosis of dense-core granules in the PC12 cells. Frequenin exhibits these biological activities through its Ca(2+) myristoyl switch, yet the switch is functionally inactive. These structural and functional traits of frequenin have been derived through the use of recombinant frequenin. In the present study, frequenin (BovFrq) native to the bovine hippocampus has been purified, sequenced for its 9 internal fragments, cloned, and studied. The findings show that structure of the BovFrq is identical to its form present in chicken, rat, mouse and human, indicating its evolutionary conservation. Its Ca(2+) myristoyl switch is active in the hippocampus. And, BovFrq physically interacts and turns on yet undisclosed ONE-GC-like ROS-GC membrane guanylate cyclase transduction machinery in the hippocampal neurons. This makes BovFrq a new Ca(2+)-sensor modulator of a novel ROS-GC transduction machinery. The study demonstrates the presence and mechanistic features of this cyclic GMP signaling pathway in the hippocampal neurons, and also provides one more support for the evolving concept where the Ca(2+)-modulated membrane guanylate cyclase transduction machinery in its variant forms is a central operational component of all neurons.
Collapse
Affiliation(s)
- Ewa Fik-Rymarkiewicz
- Unit of Regulatory and Molecular Biology, Departments of Cell Biology and Ophthalmology, SOM and NJMS, University of Medicine and Dentistry of New Jersey, Stratford, NJ 08084, USA
| | | | | |
Collapse
|
58
|
Nakamura TY, Jeromin A, Smith G, Kurushima H, Koga H, Nakabeppu Y, Wakabayashi S, Nabekura J. Novel role of neuronal Ca2+ sensor-1 as a survival factor up-regulated in injured neurons. ACTA ACUST UNITED AC 2006; 172:1081-91. [PMID: 16549499 PMCID: PMC2063765 DOI: 10.1083/jcb.200508156] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A molecular basis of survival from neuronal injury is essential for the development of therapeutic strategy to remedy neurodegenerative disorders. In this study, we demonstrate that an EF-hand Ca2+-binding protein neuronal Ca2+ sensor-1 (NCS-1), one of the key proteins for various neuronal functions, also acts as an important survival factor. Overexpression of NCS-1 rendered cultured neurons more tolerant to cell death caused by several kinds of stressors, whereas the dominant-negative mutant (E120Q) accelerated it. In addition, NCS-1 proteins increased upon treatment with glial cell line-derived neurotrophic factor (GDNF) and mediated GDNF survival signal in an Akt (but not MAPK)-dependent manner. Furthermore, NCS-1 is significantly up-regulated in response to axotomy-induced injury in the dorsal motor nucleus of the vagus neurons of adult rats in vivo, and adenoviral overexpression of E120Q resulted in a significant loss of surviving neurons, suggesting that NCS-1 is involved in an antiapoptotic mechanism in adult motor neurons. We propose that NCS-1 is a novel survival-promoting factor up-regulated in injured neurons that mediates the GDNF survival signal via the phosphatidylinositol 3-kinase-Akt pathway.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Molecular Physiology, National Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
59
|
Bett GCL, Morales MJ, Strauss HC, Rasmusson RL. KChIP2b modulates the affinity and use-dependent block of Kv4.3 by nifedipine. Biochem Biophys Res Commun 2006; 340:1167-77. [PMID: 16414350 DOI: 10.1016/j.bbrc.2005.12.135] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Accepted: 12/20/2005] [Indexed: 10/25/2022]
Abstract
Rapidly activating Kv4 voltage-gated ion channels are found in heart, brain, and diverse other tissues including colon and uterus. Kv4.3 can co-assemble with KChIP ancillary subunits, which modify kinetic behavior. We examined the affinity and use dependence of nifedipine block on Kv4.3 and its modulation by KChIP2b. Nifedipine (150 microM) reduced peak Kv4.3 current approximately 50%, but Kv4.3/KChIP2b current only approximately 27%. Nifedipine produced a very rapid component of open channel block in both Kv4.3 and Kv4.3/KChIP2b. However, recovery from the blocked/inactivated state was strongly sensitive to KChIP2b. Kv4.3 Thalf,recovery was slowed significantly by nifedipine (120.0+/-12.4 ms vs. 213.1+/-18.2 ms), whereas KChIP2b eliminated nifedipine's effect on recovery: Kv4.3/KChIP2b Thalf,recovery was 45.3+/-7.2 ms (control) and 47.8+/-8.2 ms (nifedipine). Consequently, Kv4.3 exhibited use-dependent nifedipine block in response to a series of depolarizing pulses which was abolished by KChIP2b. KChIPs alter drug affinity and use dependence of Kv4.3.
Collapse
Affiliation(s)
- Glenna C L Bett
- Center for Cellular and Systems Electrophysiology, Department of Gynecology and Obstetrics, School of Medicine and Biomedical Sciences, 124 Sherman Hall, State University of New York at Buffalo, Buffalo, NY 14214-300, USA
| | | | | | | |
Collapse
|
60
|
Ikura M, Ames JB. Genetic polymorphism and protein conformational plasticity in the calmodulin superfamily: two ways to promote multifunctionality. Proc Natl Acad Sci U S A 2006; 103:1159-64. [PMID: 16432210 PMCID: PMC1360552 DOI: 10.1073/pnas.0508640103] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Calcium signaling pathways control a variety of cellular events such as gene transcription, protein phosphorylation, nucleotide metabolism, and ion transport. These pathways often involve a large number of calcium-binding proteins collectively known as the calmodulin or EF-hand protein superfamily. Many EF-hand proteins undergo a large conformational change upon binding to Ca(2+) and target proteins. All members of the superfamily share marked sequence homology and similar structural features required to sense Ca(2+). Despite such structural similarities, the functional diversity of EF-hand calcium-binding proteins is extraordinary. Calmodulin itself can bind >300 different proteins, and the many members of the neuronal calcium sensor and S100 protein families collectively recognize a largely different set of target proteins. Recent biochemical and structural studies of many different EF-hand proteins highlight remarkable similarities and variations in conformational responses to the common ligand Ca(2+) and their respective cellular targets. In this review, we examine the essence of molecular recognition activities and the mechanisms by which calmodulin superfamily proteins control a wide variety of Ca(2+) signaling processes.
Collapse
Affiliation(s)
- Mitsuhiko Ikura
- Division of Signaling Biology, Ontario Cancer Institute and Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, Ontario, Canada M5G 2M9.
| | | |
Collapse
|
61
|
Dhar-Chowdhury P, Harrell MD, Han SY, Jankowska D, Parachuru L, Morrissey A, Srivastava S, Liu W, Malester B, Yoshida H, Coetzee WA. The glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase, triose-phosphate isomerase, and pyruvate kinase are components of the K(ATP) channel macromolecular complex and regulate its function. J Biol Chem 2005; 280:38464-70. [PMID: 16170200 PMCID: PMC4667781 DOI: 10.1074/jbc.m508744200] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of ATP-sensitive potassium (K(ATP)) channel activity is complex and a multitude of factors determine their open probability. Physiologically and pathophysiologically, the most important of these are intracellular nucleotides, with a long-recognized role for glycolytically derived ATP in regulating channel activity. To identify novel regulatory subunits of the K(ATP) channel complex, we performed a two-hybrid protein-protein interaction screen, using as bait the mouse Kir6.2 C terminus. Screening a rat heart cDNA library, we identified two potential interacting proteins to be the glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and triose-phosphate isomerase. The veracity of interaction was verified by co-immunoprecipitation techniques in transfected mammalian cells. We additionally demonstrated that pyruvate kinase also interacts with Kir6.2 subunits. The physiological relevance of these interactions is illustrated by the demonstration that native Kir6.2 protein similarly interact with GAPDH and pyruvate kinase in rat heart membrane fractions and that Kir6.2 protein co-localize with these glycolytic enzymes in rat ventricular myocytes. The functional relevance of our findings is demonstrated by the ability of GAPDH or pyruvate kinase substrates to directly block the K(ATP) channel under patch clamp recording conditions. Taken together, our data provide direct evidence for the concept that key enzymes involved in glycolytic ATP production are part of a multisubunit K(ATP) channel protein complex. Our data are consistent with the concept that the activity of these enzymes (possibly by ATP formation in the immediate intracellular microenvironment of this macromolecular K(ATP) channel complex) causes channel closure.
Collapse
Affiliation(s)
- Piyali Dhar-Chowdhury
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Maddison D. Harrell
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Sandra Y. Han
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Danuta Jankowska
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Lavanya Parachuru
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Alison Morrissey
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Shekhar Srivastava
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Weixia Liu
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Brian Malester
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Hidetada Yoshida
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - William A. Coetzee
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
62
|
Jinno S, Jeromin A, Kosaka T. Postsynaptic and extrasynaptic localization of Kv4.2 channels in the mouse hippocampal region, with special reference to targeted clustering at gabaergic synapses. Neuroscience 2005; 134:483-94. [PMID: 16009497 DOI: 10.1016/j.neuroscience.2005.04.065] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Revised: 04/09/2005] [Accepted: 04/14/2005] [Indexed: 10/25/2022]
Abstract
Voltage-dependent potassium (Kv) channels in the CNS are involved in regulation of subthreshold membrane potentials, and thus reception and integration of synaptic signals. Although such features are particularly important for induction of hippocampal synaptic plasticity, relatively little is known about their subcellular localization. Here we analyzed the detailed distribution of Kv4.2 potassium channels in the mouse hippocampal region using confocal and electron microscopy. At the light microscopic level, the Kv4.2 immunoreactivity occurred in a punctate fashion in the whole area of the hippocampal region. In the hippocampus proper, most of the Kv4.2-positive puncta were small, and they were abundant at the dendritic compartments of pyramidal neurons. High-resolution confocal microscopy revealed that there was no apparent association between Kv4.2-positive puncta with major synaptic markers, such as vesicular glutamate transporters and glutamic acid decarboxylase. In the subicular complex and dentate gyrus, we encountered large distinct Kv4.2-positive puncta at the perimeter of somata and proximal dendrites of principal cells. These puncta were often in contact with glutamic acid decarboxylase-positive boutons, but showed no apparent association with vesicular glutamate transporters. The glutamic acid decarboxylase-positive boutons apposing to Kv4.2-positive puncta were parvalbumin-positive. Quantitative image analysis showed that approximately half of Kv4.2-positive puncta were closely apposed to glutamic acid decarboxylase-positive boutons in the parasubiculum and dentate gyrus. Electron microscopic examination substantiated the presence of large Kv4.2-positive patches at postsynaptic sites of symmetric synapses and small patches at extrasynaptic sites. No presynaptic terminals were labeled. The present findings indicate targeted clustering of Kv4.2 potassium channels at postsynaptic sites of GABAergic synapses and extrasynaptic sites, and provide some key to understand their role in the hippocampal region.
Collapse
Affiliation(s)
- S Jinno
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | | | | |
Collapse
|
63
|
Patel SP, Campbell DL. Transient outward potassium current, 'Ito', phenotypes in the mammalian left ventricle: underlying molecular, cellular and biophysical mechanisms. J Physiol 2005; 569:7-39. [PMID: 15831535 PMCID: PMC1464208 DOI: 10.1113/jphysiol.2005.086223] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/07/2005] [Accepted: 04/13/2005] [Indexed: 11/08/2022] Open
Abstract
At least two functionally distinct transient outward K(+) current (I(to)) phenotypes can exist across the free wall of the left ventricle (LV). Based upon their voltage-dependent kinetics of recovery from inactivation, these two phenotypes are designated 'I(to,fast)' (recovery time constants on the order of tens of milliseconds) and 'I(to,slow)' (recovery time constants on the order of thousands of milliseconds). Depending upon species, either I(to,fast), I(to,slow) or both current phenotypes may be expressed in the LV free wall. The expression gradients of these two I(to) phenotypes across the LV free wall are typically heterogeneous and, depending upon species, may consist of functional phenotypic gradients of both I(to,fast) and I(to,slow) and/or density gradients of either phenotype. We review the present evidence (molecular, biophysical, electrophysiological and pharmacological) for Kv4.2/4.3 alpha subunits underlying LV I(to,fast) and Kv1.4 alpha subunits underlying LV I(to,slow) and speculate upon the potential roles of each of these currents in determining frequency-dependent action potential characteristics of LV subepicardial versus subendocardial myocytes in different species. We also review the possible functional implications of (i) ancillary subunits that regulate Kv1.4 and Kv4.2/4.3 (Kvbeta subunits, DPPs), (ii) KChIP2 isoforms, (iii) spider toxin-mediated block of Kv4.2/4.3 (Heteropoda toxins, phrixotoxins), and (iv) potential mechanisms of modulation of I(to,fast) and I(to,slow) by cellular redox state, [Ca(2)(+)](i) and kinase-mediated phosphorylation. I(to) phenotypic activation and state-dependent gating models and molecular structure-function relationships are also discussed.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, NY 14214-3078, USA.
| | | |
Collapse
|
64
|
Garcia N, Lanuza MA, Besalduch N, Santafe MM, Jeromin A, Tomas J. Localization of neuronal calcium sensor-1 at the adult and developing rat neuromuscular junction. J Neurosci Res 2005; 82:1-9. [PMID: 16088942 DOI: 10.1002/jnr.20593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuronal calcium sensor (NCS-1; frequenin) is a calcium-binding protein involved in the regulation of neurotransmission in the central and peripheral nervous systems from insects to vertebrates. This study reports the localization of NCS-1 immunoreactivity, by Western blotting and immunohistochemistry, at the adult and developing postnatal rat neuromuscular junction. Our confocal immunofluorescence results on the whole-mount muscle and on semithin cross-sections are indicative of the localization of NCS-1 to motor axon terminals. There is no evidence of immunoreactivity in the postsynaptic side of the neuromuscular junctions or teloglial Schwann cells. These results suggest that NCS-1 is involved in the formation and function of presynaptic nerve terminal part of the neuromuscular junction during synaptogenesis and in adult mammals.
Collapse
Affiliation(s)
- Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | | | | | | | | | | |
Collapse
|
65
|
Barclay JW, Morgan A, Burgoyne RD. Calcium-dependent regulation of exocytosis. Cell Calcium 2005; 38:343-53. [PMID: 16099500 DOI: 10.1016/j.ceca.2005.06.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Accepted: 06/28/2005] [Indexed: 11/30/2022]
Abstract
A rapid increase in intracellular calcium directly triggers regulated exocytosis. In addition, changes in intracellular calcium concentration can adjust the extent of exocytosis (quantal content) or the magnitude of individual release events (quantal size) in both the short- and long-term. It is generally agreed that calcium achieves this regulation via an interaction with a number of different molecular targets located at or near to the site of membrane fusion. We review here the synaptic proteins with defined calcium-binding domains and protein kinases activated by calcium, summarize what is known about their function in membrane fusion and the experimental evidence in support of their involvement in synaptic plasticity.
Collapse
Affiliation(s)
- Jeff W Barclay
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | |
Collapse
|
66
|
Zheng Q, Bobich JA, Vidugiriene J, McFadden SC, Thomas F, Roder J, Jeromin A. Neuronal calcium sensor-1 facilitates neuronal exocytosis through phosphatidylinositol 4-kinase. J Neurochem 2005; 92:442-51. [PMID: 15659215 DOI: 10.1111/j.1471-4159.2004.02897.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This work tested the theory that neuronal calcium sensor-1 (NCS-1) has effects on neurotransmitter release beyond its actions on membrane channels. We used nerve-ending preparations where membrane channels are bypassed through membrane permeabilization made by mechanical disruption or streptolysin-O. Nerve ending NCS-1 and phosphatidylinositol 4-kinase (PI4K) are largely or entirely particulate, so their concentrations in nerve endings remain constant after breaching the membrane. Exogenous, myristoylated NCS-1 stimulated nerve ending phosphatidylinositol 4-phosphate [PI(4)P] synthesis, but non-myristoylated-NCS-1 did not. The N-terminal peptide of NCS-1 interfered with PI(4)P synthesis, and with spontaneous and Ca(2+)-evoked release of both [(3)H]-norepinephrine (NA) and [(14)C]-glutamate (glu) in a concentration-dependent manner. An antibody raised against the N-terminal of NCS-1 inhibited perforated nerve ending PI(4)P synthesis, but the C-terminal antibody had no effects. Antibodies against the N- and C-termini of NCS-1 caused significant increases in mini/spontaneous/stimulation-independent release of [(3)H]-NA from perforated nerve endings, but had no effect on [(14)C]-glu release. These results support the idea that NCS-1 facilitates nerve ending neurotransmitter release and phosphoinositide production via PI4K and localizes these effects to the N-terminal of NCS-1. Combined with previous work on the regulation of channels by NCS-1, the data are consistent with the hypothesis that a NCS-1-PI4K (NP, neuropotentiator) complex may serve as an essential linker between lipid and protein metabolism to regulate membrane traffic and co-ordinate it with ion fluxes and plasticity in the nerve ending.
Collapse
Affiliation(s)
- Qian Zheng
- Department of Chemistry, Texas Christian University, Fort Worth, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
67
|
Bekar LK, Loewen ME, Cao K, Sun X, Leis J, Wang R, Forsyth GW, Walz W. Complex expression and localization of inactivating Kv channels in cultured hippocampal astrocytes. J Neurophysiol 2005; 93:1699-709. [PMID: 15738276 DOI: 10.1152/jn.00850.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Voltage-gated potassium channels are well established as critical for setting action potential frequency, membrane potential, and neurotransmitter release in neurons. However, their role in the "nonexcitable" glial cell type is yet to be fully understood. We used whole cell current kinetics, pharmacology, immunocytochemistry, and RT-PCR to characterize A-type current in hippocampal astrocyte cultures to better understand its function. Pharmacological analysis suggests that approximately 70, 10, and <5% of total A current is associated with Kv4, Kv3, and Kv1 channels, respectively. In addition, pharmacology and kinetics provide evidence for a significant contribution of KChIP accessory proteins to astrocytic A-channel composition. Localization of the Shaw Kv3.4 channel to astrocytic processes and the Shal Kv4.3 channel to soma suggest that these channels serve a specific function. Given this complex A-type channel expression pattern, we assessed the role of A currents in membrane voltage oscillations in response to current injections. Although TEA-sensitive delayed-rectifying currents are involved in the extent of repolarization, 4-AP-sensitive A currents serve to increase the rate. As in neurons, this effect may enable astrocytes to respond rapidly to high-frequency synaptic events. Our results indicate that hippocampal astrocytes in vitro express multiple A-type Kv channel alpha-subunits with accessory, possibly Ca(2+)-sensitive, cytoplasmic subunits that appear to be specifically localized to subcellular membrane compartments. Function of these channels remains to be determined in a physiological setting. However, this study suggests that they enable astrocytes to respond rapidly with membrane voltage oscillations to high-frequency incoming signals, possibly synchronizing astrocyte function to neuronal activity.
Collapse
MESH Headings
- 4-Aminopyridine/pharmacology
- 5,8,11,14-Eicosatetraynoic Acid/pharmacology
- Animals
- Animals, Newborn
- Astrocytes/drug effects
- Astrocytes/metabolism
- Blotting, Northern/methods
- Calcium/metabolism
- Cells, Cultured
- Electric Stimulation/methods
- Glial Fibrillary Acidic Protein/metabolism
- Hippocampus/cytology
- Hippocampus/metabolism
- Humans
- Immunohistochemistry/methods
- Ion Channel Gating/drug effects
- Ion Channel Gating/physiology
- Ion Channel Gating/radiation effects
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Membrane Potentials/radiation effects
- Patch-Clamp Techniques/methods
- Potassium Channel Blockers/pharmacology
- Potassium Channels, Voltage-Gated/classification
- Potassium Channels, Voltage-Gated/genetics
- Potassium Channels, Voltage-Gated/metabolism
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Tetraethylammonium/pharmacology
- Transfection/methods
Collapse
Affiliation(s)
- Lane K Bekar
- Department of Physiology, University of Saskatchewan, Rm B39 Health Sciences Bldg, Saskatoon, SK, S7N 5E5, Canada
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Jerng HH, Pfaffinger PJ, Covarrubias M. Molecular physiology and modulation of somatodendritic A-type potassium channels. Mol Cell Neurosci 2005; 27:343-69. [PMID: 15555915 DOI: 10.1016/j.mcn.2004.06.011] [Citation(s) in RCA: 239] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2004] [Revised: 05/22/2004] [Accepted: 06/08/2004] [Indexed: 11/23/2022] Open
Abstract
The somatodendritic subthreshold A-type K+ current (ISA) in nerve cells is a critical component of the ensemble of voltage-gated ionic currents that determine somatodendritic signal integration. The underlying K+ channel belongs to the Shal subfamily of voltage-gated K+ channels. Most Shal channels across the animal kingdom share a high degree of structural conservation, operate in the subthreshold range of membrane potentials, and exhibit relatively fast inactivation and recovery from inactivation. Mammalian Shal K+ channels (Kv4) undergo preferential closed-state inactivation with features that are generally inconsistent with the classical mechanisms of inactivation typical of Shaker K+ channels. Here, we review (1) the physiological and genetic properties of ISA, 2 the molecular mechanisms of Kv4 inactivation and its remodeling by a family of soluble calcium-binding proteins (KChIPs) and a membrane-bound dipeptidase-like protein (DPPX), and (3) the modulation of Kv4 channels by protein phosphorylation.
Collapse
Affiliation(s)
- Henry H Jerng
- Division of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | |
Collapse
|
69
|
Duzhyy D, Harvey M, Sokolowski B. A secretory-type protein, containing a pentraxin domain, interacts with an A-type K+ channel. J Biol Chem 2005; 280:15165-72. [PMID: 15708850 DOI: 10.1074/jbc.m500111200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A-type K(+) channels belonging to the Shal subfamily are found in various receptor and neuronal cells. Although their kinetics and cell surface expression are regulated by auxiliary subunits, little is known about the proteins that may interact with Kv4 during development. A yeast two-hybrid screening of a cDNA library made from the sensory epithelium of embryonic chick cochlea revealed a novel association of Kv4.2 with a protein containing a pentraxin domain (PPTX). Sequence analysis shows that PPTX is a member of the long pentraxin family, is 53% identical to mouse PTX3, and has a signal peptide at the N terminus. Studies with chick cochlear tissues reveal that Kv4.2 coprecipitates PPTX and that both proteins are colocalized to the sensory and ganglion cells. A yeast two-hybrid assay demonstrated that the last 22 amino acids of the PPTX C terminus interact with the N terminus of Kv4.2. Chinese hamster ovary cells transfected with recombinant PPTX reveal secretory products in both non-truncated and truncated forms. Among the secreted variants are several blocked by Brefeldin A, suggesting export via a classical pathway. PPTX is soluble in the presence of sodium carbonate, suggesting localization to the cytosolic side of the plasmalemma. Immunohistochemical studies show that Kv4.2 and PPTX colocalize in the region of the plasmalemma of Chinese hamster ovary cells; however, both are locked in the endoplasmic reticulum of COS-7 cells, suggesting that PPTX does not act as a shuttle protein. Reverse transcription-PCR demonstrates that PPTX mRNA is found in tissues that include brain, eye, heart, and blood vessels.
Collapse
Affiliation(s)
- Dmytro Duzhyy
- University of South Florida, Department of Otolaryngology, Head and Neck Surgery, Tampa, Florida 33612, USA
| | | | | |
Collapse
|
70
|
D Burgoyne R. The neuronal calcium-sensor proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2004; 1742:59-68. [PMID: 15590056 DOI: 10.1016/j.bbamcr.2004.08.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Accepted: 08/16/2004] [Indexed: 10/26/2022]
Abstract
Changes in intracellular free Ca(2+) concentration ([Ca(2+)](i)) affect many different aspects of neuronal function ranging from millisecond regulation of ion channels to long term changes in gene expression. These effects of Ca(2+) are transduced by Ca(2+)-binding proteins that act as Ca(2+) sensors by binding Ca(2+), undergoing a conformational change and then modifying the function of additional target proteins. Mammalian species express 14 members of the neuronal calcium sensor (NCS) family of EF hand-containing Ca(2+)-binding proteins which are expressed mainly in photoreceptor cells or neurons. Many of the NCS proteins are membrane targeted through their N-terminal myristoylation either constitutively or following exposure of the myristoyl group after Ca(2+) binding (the Ca(2+)/myristoyl switch). The NCS proteins have been implicated in a wide range of functional roles in neuronal regulation, several of which have been confirmed though molecular genetic analyses.
Collapse
Affiliation(s)
- Robert D Burgoyne
- The Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| |
Collapse
|
71
|
Brackmann M, Zhao C, Kuhl D, Manahan-Vaughan D, Braunewell KH. MGluRs regulate the expression of neuronal calcium sensor proteins NCS-1 and VILIP-1 and the immediate early gene arg3.1/arc in the hippocampus in vivo. Biochem Biophys Res Commun 2004; 322:1073-9. [PMID: 15336574 DOI: 10.1016/j.bbrc.2004.08.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2004] [Indexed: 10/26/2022]
Abstract
The metabotropic glutamate receptor (mGluR) agonist (R,S)-3,5-dihydroxyphenylglycine (DHPG) is involved in several forms of hippocampal synaptic plasticity. DHPG application can induce slow-onset potentiation, a form of long-term potentiation (LTP), in the dentate gyrus and in the CA1 region in vivo. The induction of LTP correlates with increased expression levels of neuronal calcium sensor (NCS), considered as key elements for plasticity. In this study we investigated mGluR- and time-dependent changes in the expression of two different NCS proteins. Following DHPG application in vivo NCS-1 and VILIP-1 expression increased, with significant levels reached after 8 and 24h. The effect was attenuated by treatment with the group I mGluR specific antagonist S-4-carboxyphenylglycine. The immediate early gene (IEG) arg3.1/arc showed highest expression levels 2h after DHPG-treatment. Therefore, mGluRs at concentrations which induce synaptic plasticity regulate the expression of IEGs and NCS proteins in different time frames and thus contribute to late phases of synaptic plasticity.
Collapse
Affiliation(s)
- Marian Brackmann
- Signal Transduction Research Group, Neuroscience Research Center of the Charite, Tucholskystrasse 2, D-10117 Berlin, Germany
| | | | | | | | | |
Collapse
|
72
|
Bogdanik L, Mohrmann R, Ramaekers A, Bockaert J, Grau Y, Broadie K, Parmentier ML. The Drosophila metabotropic glutamate receptor DmGluRA regulates activity-dependent synaptic facilitation and fine synaptic morphology. J Neurosci 2004; 24:9105-16. [PMID: 15483129 PMCID: PMC6730051 DOI: 10.1523/jneurosci.2724-04.2004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2004] [Revised: 08/02/2004] [Accepted: 08/03/2004] [Indexed: 11/21/2022] Open
Abstract
In vertebrates, several groups of metabotropic glutamate receptors (mGluRs) are known to modulate synaptic properties. In contrast, the Drosophila genome encodes a single functional mGluR (DmGluRA), an ortholog of vertebrate group II mGluRs, greatly expediting the functional characterization of mGluR-mediated signaling in the nervous system. We show here that DmGluRA is expressed at the glutamatergic neuromuscular junction (NMJ), localized in periactive zones of presynaptic boutons but excluded from active sites. Null DmGluRA mutants are completely viable, and all of the basal NMJ synaptic transmission properties are normal. In contrast, DmGluRA mutants display approximately a threefold increase in synaptic facilitation during short stimulus trains. Prolonged stimulus trains result in very strongly increased ( approximately 10-fold) augmentation, including the appearance of asynchronous, bursting excitatory currents never observed in wild type. Both defects are rescued by expression of DmGluRA only in the neurons, indicating a specific presynaptic requirement. These phenotypes are reminiscent of hyperexcitable mutants, suggesting a role of DmGluRA signaling in the regulation of presynaptic excitability properties. The mutant phenotypes could not be replicated by acute application of mGluR antagonists, suggesting that DmGluRA regulates the development of presynaptic properties rather than directly controlling short-term modulation. DmGluRA mutants also display mild defects in NMJ architecture: a decreased number of synaptic boutons accompanied by an increase in mean bouton size. These morphological changes bidirectionally correlate with DmGluRA levels in the presynaptic terminal. These data reveal the following two roles for DmGluRA in presynaptic mechanisms: (1) modulation of presynaptic excitability properties important for the control of activity-dependent neurotransmitter release and (2) modulation of synaptic architecture.
Collapse
Affiliation(s)
- Laurent Bogdanik
- Laboratoire de Génomique Fonctionnelle, Centre National de la Recherche Scientifique, Unité Propre de Recherche 2580, 34094 Montpellier Cedex 05, France
| | | | | | | | | | | | | |
Collapse
|
73
|
Baranauskas G. Cell-type-specific splicing of KChIP4 mRNA correlates with slower kinetics of A-type current. Eur J Neurosci 2004; 20:385-91. [PMID: 15233748 DOI: 10.1111/j.1460-9568.2004.03494.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In neurons, rapidly inactivating A-type potassium currents regulate repetitive firing and sensitivity to synaptic inputs both in the soma and in the dendrites. It has been established that Kv4 family subunits with several modifying proteins such as KChIPs are responsible for A-type current in most neurons. However, it is not clear which of these modifying proteins are responsible for the observed difference in the properties of A-type currents in the neurons. For example, in globus pallidus (GP) and basal forebrain (BF) neurons in rats, A-type current possesses a slowly inactivating (tau > 80 ms) component of inactivation that is absent in the currents obtained from striatal cholinergic interneurons (StrI) and hippocampal area CA1 pyramidal neurons (HIP). It has been shown that KChIP4 splice variant A but not splice variant B can increase inactivation rates of Kv4 current to > 100 ms in Xenopus oocytes. We tested the hypothesis that cell-specific expression of KChIP4A is responsible for the slow inactivation of A-type current in these neurons. Employing single-cell RT-PCR in acutely dissociated rat neurons, KChIP4A mRNA was detected in 12/14 GP cells and in 12/14 BF neurons whereas it was not detected in any StrI or HIP cells. By contrast, the KChIP4 splice variant B was detected in all four types of cells. Moreover, deactivation rates at -100 mV were slower in BF and GP cells than in StrI and HIP neurons as expected, owing to the presence KChIP4A in BF and GP neurons. These data are consistent with our initial hypothesis.
Collapse
Affiliation(s)
- Gytis Baranauskas
- Department of Psychiatry, Psychiatric Institute, 1601 W. Taylor St., Room 330 W, Chicago, IL 60612, USA.
| |
Collapse
|
74
|
Birnbaum SG, Varga AW, Yuan LL, Anderson AE, Sweatt JD, Schrader LA. Structure and function of Kv4-family transient potassium channels. Physiol Rev 2004; 84:803-33. [PMID: 15269337 DOI: 10.1152/physrev.00039.2003] [Citation(s) in RCA: 268] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Shal-type (Kv4.x) K(+) channels are expressed in a variety of tissue, with particularly high levels in the brain and heart. These channels are the primary subunits that contribute to transient, voltage-dependent K(+) currents in the nervous system (A currents) and the heart (transient outward current). Recent studies have revealed an enormous degree of complexity in the regulation of these channels. In this review, we describe the surprisingly large number of ancillary subunits and scaffolding proteins that can interact with the primary subunits, resulting in alterations in channel trafficking and kinetic properties. Furthermore, we discuss posttranslational modification of Kv4.x channel function with an emphasis on the role of kinase modulation of these channels in regulating membrane properties. This concept is especially intriguing as Kv4.2 channels may integrate a variety of intracellular signaling cascades into a coordinated output that dynamically modulates membrane excitability. Finally, the pathophysiology that may arise from dysregulation of these channels is also reviewed.
Collapse
Affiliation(s)
- Shari G Birnbaum
- Div. of Neuroscience, S607, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
75
|
Burgoyne RD, O'Callaghan DW, Hasdemir B, Haynes LP, Tepikin AV. Neuronal Ca2+-sensor proteins: multitalented regulators of neuronal function. Trends Neurosci 2004; 27:203-9. [PMID: 15046879 DOI: 10.1016/j.tins.2004.01.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Many aspects of neuronal activity are regulated by Ca2+ signals. The transduction of temporally and spatially distinct Ca2+ signals requires the action of Ca2+-sensor proteins including various EF-hand-containing Ca2+-binding proteins. The neuronal Ca2+ sensor (NCS) protein family and the related Ca2+-binding proteins (CaBPs) have begun to emerge as key players in neuronal function. Many of these proteins are expressed predominantly or only in neurons, sometimes with cell-specific patterns of expression. Their ability to associate with membranes either constitutively or in response to elevated Ca2+ concentration allows the NCS proteins to discriminate between different spatial and temporal patterns of Ca2+ signals. Recent work has established several physiological roles of these proteins, including diverse actions on gene expression, ion channel function, membrane traffic of ion channels and receptors, and the control of apoptosis.
Collapse
Affiliation(s)
- Robert D Burgoyne
- The Physiological Laboratory, University of Liverpool, Crown Street, Liverpool L69 3BX, UK.
| | | | | | | | | |
Collapse
|
76
|
O'Callaghan DW, Hasdemir B, Leighton M, Burgoyne RD. Residues within the myristoylation motif determine intracellular targeting of the neuronal Ca2+ sensor protein KChIP1 to post-ER transport vesicles and traffic of Kv4 K+ channels. J Cell Sci 2004; 116:4833-45. [PMID: 14600268 DOI: 10.1242/jcs.00803] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
KChIPs (K+ channel interacting proteins) regulate the function of A-type Kv4 potassium channels by modifying channel properties and by increasing their cell surface expression. We have explored factors affecting the localisation of Kv4.2 and the targeting of KChIP1 and other NCS proteins by using GFP-variant fusion proteins expressed in HeLa cells. ECFP-Kv4.2 expressed alone was not retained in the ER but reached the Golgi complex. In cells co-expressing ECFP-Kv4.2 and KChIP1-EYFP, the two proteins were co-localised and were mainly present on the plasma membrane. When KChIP1-EYFP was expressed alone it was instead targeted to punctate structures. This was distinct from the localisation of the NCS proteins NCS-1 and hippocalcin, which were targeted to the trans-Golgi network (TGN) and plasma membrane. The membrane localisation of each NCS protein required myristoylation and minimal myristoylation motifs of hippocalcin or KChIP1 were sufficient to target fusion proteins to either TGN/plasma membrane or to punctate structures. The existence of targeting information within the N-terminal motifs was confirmed by mutagenesis of residues corresponding to three conserved basic amino acids in hippocalcin and NCS-1 at positions 3, 7 and 9. Residues at these positions determined intracellular targeting to the different organelles. Myristoylation and correct targeting of KChIP1 was required for the efficient traffic of ECFP-Kv4.2 to the plasma membrane. Expression of KChIP1(1-11)-EYFP resulted in the formation of enlarged structures that were positive for ERGIC-53 and beta-COP. ECFP-Kv4.2 was also accumulated in these structures suggesting that KChIP1(1-11)-EYFP inhibited traffic out of the ERGIC. We suggest that KChIP1 is targeted by its myristoylation motif to post-ER transport vesicles where it could interact with and regulate the traffic of Kv4 channels to the plasma membrane under the influence of localised Ca2+ signals.
Collapse
Affiliation(s)
- Dermott W O'Callaghan
- The Physiological Laboratory, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | | | |
Collapse
|
77
|
Zhou W, Qian Y, Kunjilwar K, Pfaffinger PJ, Choe S. Structural insights into the functional interaction of KChIP1 with Shal-type K(+) channels. Neuron 2004; 41:573-86. [PMID: 14980206 DOI: 10.1016/s0896-6273(04)00045-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2003] [Revised: 10/21/2003] [Accepted: 12/16/2003] [Indexed: 11/24/2022]
Abstract
Four Kv channel-interacting proteins (KChIP1 through KChIP4) interact directly with the N-terminal domain of three Shal-type voltage-gated potassium channels (Kv4.1, Kv4.2, and Kv4.3) to modulate cell surface expression and function of Kv4 channels. Here we report a 2.0 Angstrom crystal structure of the core domain of KChIP1 (KChIP1*) in complex with the N-terminal fragment of Kv4.2 (Kv4.2N30). The complex reveals a clam-shaped dimeric assembly. Four EF-hands from each KChIP1 form each shell of the clam. The N-terminal end of Kv4.2 forming an alpha helix (alpha1) and the C-terminal alpha helix (H10) of KChIP1 are enclosed nearly coaxially by these shells. As a result, the H10 of KChIP1 and alpha1 of Kv4.2 mediate interactions between these two molecules, structurally reminiscent of the interactions between calmodulin and its target peptides. Site-specific mutagenesis combined with functional characterization shows that those interactions mediated by alpha1 and H10 are essential to the modulation of Kv4.2 by KChIPs.
Collapse
Affiliation(s)
- Wei Zhou
- Division of Neuroscience, Baylor College of Medicine, Houston, TX 77030 USA
| | | | | | | | | |
Collapse
|
78
|
Kim LA, Furst J, Gutierrez D, Butler MH, Xu S, Goldstein SAN, Grigorieff N. Three-dimensional structure of I(to); Kv4.2-KChIP2 ion channels by electron microscopy at 21 Angstrom resolution. Neuron 2004; 41:513-9. [PMID: 14980201 DOI: 10.1016/s0896-6273(04)00050-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2003] [Revised: 01/16/2004] [Accepted: 01/22/2004] [Indexed: 01/16/2023]
Abstract
Regulatory KChIP2 subunits assemble with pore-forming Kv4.2 subunits in 4:4 complexes to produce native voltage-gated potassium (Kv) channels like cardiac I(to) and neuronal I(A) subtypes. Here, negative stain electron microscopy (EM) and single particle averaging reveal KChIP2 to create a novel approximately 35 x 115 x 115 Angstrom, intracellular fenestrated rotunda: four peripheral columns that extend down from the membrane-embedded portion of the channel to enclose the Kv4.2 "hanging gondola" (a platform held beneath the transmembrane conduction pore by four internal columns). To reach the pore from the cytosol, ions traverse one of four external fenestrae to enter the rotundal vestibule and then cross one of four internal windows in the gondola.
Collapse
Affiliation(s)
- Leo A Kim
- Department of Pediatrics, Boyer Center for Molecular Medicine, Yale University School of Medicine, 295 Congress Avenue, New Haven, CT 06535, USA
| | | | | | | | | | | | | |
Collapse
|
79
|
Patel SP, Parai R, Parai R, Campbell DL. Regulation of Kv4.3 voltage-dependent gating kinetics by KChIP2 isoforms. J Physiol 2004; 557:19-41. [PMID: 14724186 PMCID: PMC1665034 DOI: 10.1113/jphysiol.2003.058172] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We conducted a kinetic analysis of the voltage dependence of macroscopic inactivation (tau(fast), tau(slow)), closed-state inactivation (tau(closed,inact)), recovery (tau(rec)), activation (tau(act)), and deactivation (tau(deact)) of Kv4.3 channels expressed alone in Xenopus oocytes and in the presence of the calcium-binding ancillary subunits KChIP2b and KChIP2d. We demonstrate that for all expression conditions, tau(rec), tau(closed,inact) and tau(fast) are components of closed-state inactivation transitions. The values of tau(closed,inact) and tau(fast) monotonically merge from -30 to -20 mV while the values of tau(closed,inact) and tau(rec) approach each other from -60 to -50 mV. These data generate classic bell-shaped time-constant-potential curves. With the KChIPs, these curves are distinct from that of Kv4.3 expressed alone due to acceleration of tau(rec) and slowing of tau(closed,inact) and tau(fast). Only at depolarized potentials where channels open is tau(slow) detectable suggesting that it represents an open-state inactivation mechanism. With increasing depolarization, KChIPs favour this open-state inactivation mechanism, supported by the observation of larger transient reopening currents upon membrane hyperpolarization compared to Kv4.3 expressed alone. We propose a Kv4.3 gating model wherein KChIP2 isoforms accelerate recovery, slow closed-state inactivation, and promote open-state inactivation. This model supports the observations that with KChIPs, closed-state inactivation transitions are [Ca(2+)](i)-independent, while open-state inactivation is [Ca(2+)](i)-dependent. The selective KChIP- and Ca(2+)-dependent modulation of Kv4.3 inactivation mechanisms predicted by this model provides a basis for dynamic modulation of the native cardiac transient outward current by intracellular Ca(2+) fluxes during the action potential.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, 124 Sherman Hall, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
80
|
Averill S, Robson LG, Jeromin A, Priestley JV. Neuronal calcium sensor-1 is expressed by dorsal root ganglion cells, is axonally transported to central and peripheral terminals, and is concentrated at nodes. Neuroscience 2004; 123:419-27. [PMID: 14698749 DOI: 10.1016/j.neuroscience.2003.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Neuronal calcium sensor-1 (NCS-1) is a member of the EF-hand calcium-binding protein superfamily which has been implicated in the modulation of a number of neuronal functions. In this study we have examined the expression of NCS-1 in adult rat dorsal root ganglion (DRG) neurons. NCS-1 immunoreactivity was present in most DRG neurons, including many calcitonin gene-related peptide (CGRP) expressing ones. NCS-1 showed some colocalization with the synaptic vesicle protein synaptophysin and underwent both anterograde and retrograde axonal transport. NCS-1 immunoreactivity was also present in the dorsal horn of the spinal cord, and in peripheral cutaneous terminals innervating blood vessels, where it was coexpressed with CGRP. In addition, NCS-1 in peripheral nerves was concentrated at nodes and adjoining paranodes. These results suggest novel roles for NCS-1, particularly in relation to channel function at nodes and to the peripheral release of vasoactive peptides.
Collapse
Affiliation(s)
- S Averill
- Neuroscience Centre, St. Bartholomew's and the Royal London School of Medicine and Dentistry, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | | | | | | |
Collapse
|
81
|
Kim LA, Furst J, Butler MH, Xu S, Grigorieff N, Goldstein SAN. Ito channels are octomeric complexes with four subunits of each Kv4.2 and K+ channel-interacting protein 2. J Biol Chem 2003; 279:5549-54. [PMID: 14623880 DOI: 10.1074/jbc.m311332200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mammalian voltage-gated K+ channels are assemblies of pore-forming alpha-subunits and modulating beta-subunits. To operate correctly, Kv4 alpha-subunits in the heart and central nervous system require recently identified beta-subunits of the neuronal calcium sensing protein family called K+ channel-interacting proteins (KChIPs). Here, Kv4.2.KChIP2 channels are purified, integrity of isolated complexes confirmed, molar ratio of the subunits determined, and subunit valence established. A complex has 4 subunits of each type, a stoichiometry expected for other channels employing neuronal calcium sensing beta-subunits.
Collapse
Affiliation(s)
- Leo A Kim
- Department of Pediatrics, Boyer Center for Molecular Medicine, Yale University School of Medicine, New Haven, Connecticut 06536, USA
| | | | | | | | | | | |
Collapse
|
82
|
Kapp-Barnea Y, Melnikov S, Shefler I, Jeromin A, Sagi-Eisenberg R. Neuronal Calcium Sensor-1 and Phosphatidylinositol 4-Kinase β Regulate IgE Receptor-Triggered Exocytosis in Cultured Mast Cells. THE JOURNAL OF IMMUNOLOGY 2003; 171:5320-7. [PMID: 14607934 DOI: 10.4049/jimmunol.171.10.5320] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We examined the possible occurrence and function of neuronal Ca(2+) sensor 1 (NCS-1/frequenin) in the mast cell line rat basophilic leukemia, RBL-2H3. This protein has been implicated in the control of neurosecretion from dense core granules in neuronal cells as well as in the control of constitutive secretory pathways in both yeast and mammalian cells. We show that RBL-2H3 cells, secretory cells of the immune system, endogenously express the 22-kDa NCS-1 protein as well as an immune-related 50-kDa protein. Both proteins associate in vivo with phosphatidylinositol 4-kinase beta (PI4Kbeta) and colocalize with the enzyme in the Golgi region. We show further that overexpression of NCS-1 in RBL-2H3 cells stimulates the catalytic activity of PI4Kbeta, increases IgE receptor (FcepsilonRI)-triggered hydrolysis of phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)), and stimulates FcepsilonRI-triggered, but not Ca(2+) ionophore-triggered, exocytosis. Conversely, expression of a kinase-dead mutant of PI4Kbeta reduces PI4Kbeta activity, decreases FcepsilonRI-stimulated phosphatidylinositol 4,5-bisphosphate hydrolysis, and blocks FcepsilonRI-triggered, but not Ca(2+) ionophore-triggered, exocytosis. Our results indicate that PI(4)P, produced by the Golgi-localized PI4Kbeta, is the rate-limiting factor in the synthesis of the pool of PI(4,5)P(2) that serves as substrate for the generation of lipid-derived second messengers in FcepsilonRI-triggered cells. We conclude that NCS-1 is involved in the control of regulated exocytosis in nonneural cells, where it contributes to stimulus-secretion coupling by interacting with PI4Kbeta and positive regulation of its activity.
Collapse
Affiliation(s)
- Yaara Kapp-Barnea
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
83
|
Abstract
Calsenilin has been identified as a presenilin-binding protein, a transcription factor regulating dynorphin expression, and a beta-subunit of Kv4 channels and could, thus, be a multifunctional protein. To study these functions of calsenilin in vivo and to determine the neuroanatomical expression pattern of calsenilin, we generated mice with a disruption of the calsenilin gene by the targeted insertion of the beta-galactosidase gene. We found that calsenilin expression (as represented by beta-galactosidase activity) is very restricted but overlaps better with that of presenilins and Kv4 channels than with dynorphin, suggesting that calsenilin may regulate presenilin and Kv4 channels in brain. Abeta peptide levels are reduced in calsenilin knock-out mice, demonstrating that calsenilin affects presenilin-dependent gamma-cleavage in vivo. Furthermore, long-term potentiation (LTP) in dentate gyrus of hippocampus, in which calsenilin is strongly and selectively expressed, is enhanced in calsenilin knock-out mice. This enhancement of LTP coincides with a downregulation of the Kv4 channel-dependent A-type current and can be mimicked in wild-type animals by a Kv4 channel blocker. The data presented here show that lack of calsenilin affects both Abeta formation and the A-type current. We suggest that these effects are separate events, caused by a common mechanism possibly involving protein transport.
Collapse
|
84
|
Spilker C, Braunewell KH. Calcium–myristoyl switch, subcellular localization, and calcium-dependent translocation of the neuronal calcium sensor protein VILIP-3, and comparison with VILIP-1 in hippocampal neurons☆. Mol Cell Neurosci 2003; 24:766-78. [PMID: 14664824 DOI: 10.1016/s1044-7431(03)00242-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Neuronal calcium sensor (NCS) proteins including the subfamily of visinin-like-proteins (VILIPs) are involved in regulation of various signaling cascades. One molecular regulation mechanism is the calcium-myristoyl switch. VILIPs show a calcium-dependent membrane association in brain homogenates; however, differences in calcium-induced conformation changes and degree of membrane association are reported. Little is known about differences in the calcium-myristoyl switch in living cells leading to localization of VILIPs to distinct subcellular compartments. Therefore, we studied the calcium-dependent localization of green fluorescent protein (GFP)-tagged VILIP-3 in living cell lines and hippocampal neurons and compared it with that of GFP-VILIP-1. Interestingly, the observed fast and reversible calcium-myristoyl switch of VILIP-3-GFP and VILIP-1-GFP differed, e.g., in calcium-dependent translocation to Golgi membranes. Similarily, the calcium-dependent localization of endogenously expressed VILIP-3 and -1 in dendrites differed. Thus, VILIPs co-expressed in the same neuron show clear differences in calcium-dependent localization which may allow neurons a highly selective response to various calcium stimuli.
Collapse
Affiliation(s)
- Christina Spilker
- Signal Transduction Research Group, Neuroscience Research Center/Institute for Physiology of the Charite, Humboldt University Berlin, Tucholskystrasse 2, D-10117 Berlin, Germany
| | | |
Collapse
|
85
|
Ren X, Shand SH, Takimoto K. Effective association of Kv channel-interacting proteins with Kv4 channel is mediated with their unique core peptide. J Biol Chem 2003; 278:43564-70. [PMID: 12928444 DOI: 10.1074/jbc.m302337200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Kv channel-interacting proteins (KChIPs) and neuronal calcium sensor-1 (NCS-1) have been shown to interact with Kv4 channel alpha-subunits to regulate the expression and/or gating of these channels. Here we examine the specificity and sites of these proteins for interaction with Kv channel proteins. Immunoprecipitation and green fluorescent protein imaging show that KChIPs (but not NCS-1) effectively bind to Kv4.3 protein and localize at the plasma membrane when channel proteins are coexpressed. Analysis with chimeric proteins between KChIP2 and NCS-1 reveals that the three regions of KChIP2 (the linker between the first and second EF hands, the one between the third and fourth EF hands, and the C-terminal peptide after the fourth EF hand) are necessary and sufficient for its effective binding to Kv4.3 protein. The chimera with these three KChIP2 portions slowed inactivation and facilitated recovery from inactivation of Kv4.3 current. These results indicate that the sequence difference in these three regions between KChIPs and NCS-1 determines the specificity and affinity for interaction with Kv4 protein. Because the three identified regions surround the large hydrophobic crevice based on the NCS-1 crystal structure, this crevice may be the association site of KChIPs for the channel protein.
Collapse
Affiliation(s)
- Xiaomeng Ren
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | |
Collapse
|
86
|
Lilliehook C, Bozdagi O, Yao J, Gomez-Ramirez M, Zaidi NF, Wasco W, Gandy S, Santucci AC, Haroutunian V, Huntley GW, Buxbaum JD. Altered Abeta formation and long-term potentiation in a calsenilin knock-out. J Neurosci 2003; 23:9097-106. [PMID: 14534243 PMCID: PMC6740834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023] Open
Abstract
Calsenilin has been identified as a presenilin-binding protein, a transcription factor regulating dynorphin expression, and a beta-subunit of Kv4 channels and could, thus, be a multifunctional protein. To study these functions of calsenilin in vivo and to determine the neuroanatomical expression pattern of calsenilin, we generated mice with a disruption of the calsenilin gene by the targeted insertion of the beta-galactosidase gene. We found that calsenilin expression (as represented by beta-galactosidase activity) is very restricted but overlaps better with that of presenilins and Kv4 channels than with dynorphin, suggesting that calsenilin may regulate presenilin and Kv4 channels in brain. Abeta peptide levels are reduced in calsenilin knock-out mice, demonstrating that calsenilin affects presenilin-dependent gamma-cleavage in vivo. Furthermore, long-term potentiation (LTP) in dentate gyrus of hippocampus, in which calsenilin is strongly and selectively expressed, is enhanced in calsenilin knock-out mice. This enhancement of LTP coincides with a downregulation of the Kv4 channel-dependent A-type current and can be mimicked in wild-type animals by a Kv4 channel blocker. The data presented here show that lack of calsenilin affects both Abeta formation and the A-type current. We suggest that these effects are separate events, caused by a common mechanism possibly involving protein transport.
Collapse
Affiliation(s)
- Christina Lilliehook
- Laboratory of Molecular Neuropsychiatry and Department of Psychiatry, Mount Sinai School of Medicine of New York University, New York, New York 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Strahl T, Grafelmann B, Dannenberg J, Thorner J, Pongs O. Conservation of regulatory function in calcium-binding proteins: human frequenin (neuronal calcium sensor-1) associates productively with yeast phosphatidylinositol 4-kinase isoform, Pik1. J Biol Chem 2003; 278:49589-99. [PMID: 14512421 DOI: 10.1074/jbc.m309017200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Frequenin, also known as neuronal calcium sensor-1 (NCS-1), is an N-myristoylated Ca2+-binding protein that has been conserved in both sequence and three-dimensional fold during evolution. We demonstrate using both genetic and biochemical approaches that the observed structural conservation between Saccharomyces cerevisiae frequenin (Frq1) and human NCS-1 is also reflected at the functional level. In yeast, the sole essential target of Frq1 is the phosphatidylinositol 4-kinase isoform, Pik1; both FRQ1 and PIK1 are indispensable for cell viability. Expression of human NCS-1 in yeast, but not a close relative (human KChIP2), rescues the inviability of frq1 cells. Furthermore, in vitro, Frq1 and NCS-1 (either N-myristoylated or unmyristoylated) compete for binding to a small 28-residue motif near the N terminus of Pik1. Site-directed mutagenesis indicates that the binding determinant in Pik1 is a hydrophobic alpha-helix and that frequenins bind to one side of this alpha-helix. We propose, therefore, that the function of NCS-1 in mammals may closely resemble that of Frq1 in S. cerevisiae and, hence, that frequenins in general may serve as regulators of certain isoforms of phosphatidylinositol 4-kinase.
Collapse
Affiliation(s)
- Thomas Strahl
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720-3200, USA
| | | | | | | | | |
Collapse
|
88
|
Zhang Y, MacLean JN, An WF, Lanning CC, Harris-Warrick RM. KChIP1 and frequenin modify shal-evoked potassium currents in pyloric neurons in the lobster stomatogastric ganglion. J Neurophysiol 2003; 89:1902-9. [PMID: 12612050 DOI: 10.1152/jn.00837.2002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The transient potassium current (I(A)) plays an important role in shaping the firing properties of pyloric neurons in the stomatogastric ganglion (STG) of the spiny lobster, Panulirus interruptus. The shal gene encodes I(A) in pyloric neurons. However, when we over-expressed the lobster Shal protein by shal RNA injection into the pyloric dilator (PD) neuron, the increased I(A) had somewhat different properties from the endogenous I(A). The recently cloned K-channel interacting proteins (KChIPs) can modify vertebrate Kv4 channels in cloned cell lines. When we co-expressed hKChIP1 with lobster shal in Xenopus oocytes or lobster PD neurons, they produced A-currents resembling the endogenous I(A) in PD neurons; compared with currents evoked by shal alone, their voltage for half inactivation was depolarized, their kinetics of inactivation were slowed, and their recovery from inactivation was accelerated. We also co-expressed shal in PD neurons with lobster frequenin, which encodes a protein belonging to the same EF-hand family of Ca(2+) sensing proteins as hKChIP. Frequenin also restored most of properties of the shal-evoked currents to those of the endogenous A-currents, but the time course of recovery from inactivation was not corrected. These results suggest that lobster shal proteins normally interact with proteins in the KChIP/frequenin family to produce the transient potassium current in pyloric neurons.
Collapse
Affiliation(s)
- Y Zhang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York 14853, USA.
| | | | | | | | | |
Collapse
|
89
|
Nakamura TY, Sturm E, Pountney DJ, Orenzoff B, Artman M, Coetzee WA. Developmental expression of NCS-1 (frequenin), a regulator of Kv4 K+ channels, in mouse heart. Pediatr Res 2003; 53:554-7. [PMID: 12612193 DOI: 10.1203/01.pdr.0000057203.72435.c9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The channel proteins responsible for the cardiac transient outward K+ current (Ito) of human and rodent heart are composed, in part, of pore-forming Kv4.3 or Kv4.2 principal subunits. Recent reports implicate K+ channel interacting proteins (members of the neuronal Ca2+-binding protein family) as subunits of the Ito channel complex. We reported that another Ca2+-binding protein, frequenin [or neuronal calcium center protein-1 (NCS-1)], also functions as a Kv4 auxiliary subunit in the brain. By examining cardiac expression of NCS-1, the aim of this study was to examine the potential physiologic relevance of this protein as an additional regulator of cardiac Ito. Immunoblot analysis demonstrates NCS-1 protein to be expressed in adult mouse ventricle at levels comparable to that found in some brain regions. Cardiac NCS-1 protein expression levels are much higher in fetal and neonatal mouse hearts when compared with the adult. Immunocytochemical analysis of isolated neonatal mouse ventricular myocytes demonstrates co-localization of NCS-1 and Kv4.2 proteins at the sarcolemma. Given its high levels of expression in the heart, NCS-1 should be considered an important potential Kv4 regulatory subunit, particularly in the immature heart.
Collapse
Affiliation(s)
- Tomoe Y Nakamura
- Department of Pediatric Cardiology, New York University School of Medicine, New York 10016, USA
| | | | | | | | | | | |
Collapse
|
90
|
Amberg GC, Koh SD, Imaizumi Y, Ohya S, Sanders KM. A-type potassium currents in smooth muscle. Am J Physiol Cell Physiol 2003; 284:C583-95. [PMID: 12556357 DOI: 10.1152/ajpcell.00301.2002] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A-type currents are voltage-gated, calcium-independent potassium (Kv) currents that undergo rapid activation and inactivation. Commonly associated with neuronal and cardiac cell-types, A-type currents have also been identified and characterized in vascular, genitourinary, and gastrointestinal smooth muscle cells. This review examines the molecular identity, biophysical properties, pharmacology, regulation, and physiological function of smooth muscle A-type currents. In general, this review is intended to facilitate the comparison of A-type currents present in different smooth muscles by providing a comprehensive report of the literature to date. This approach should also aid in the identification of areas of research requiring further attention.
Collapse
Affiliation(s)
- Gregory C Amberg
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno 89557, USA
| | | | | | | | | |
Collapse
|
91
|
Rousset M, Cens T, Gavarini S, Jeromin A, Charnet P. Down-regulation of voltage-gated Ca2+ channels by neuronal calcium sensor-1 is beta subunit-specific. J Biol Chem 2003; 278:7019-26. [PMID: 12496289 DOI: 10.1074/jbc.m209537200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal Ca(2+) sensor protein-1 (NCS-1) is a member of the Ca(2+) binding protein family, with three functional Ca(2+) binding EF-hands and an N-terminal myristoylation site. NCS-1 is expressed in brain and heart during embryonic and postnatal development. In neurons, NCS-1 facilitates neurotransmitter release, but both inhibition and facilitation of the Ca(2+) current amplitude have been reported. In heart, NCS-1 co-immunoprecipitates with K(+) channels and modulates their activity, but the potential effects of NCS-1 on cardiac Ca(2+) channels have not been investigated. To directly assess the effect of NCS-1 on the various types of Ca(2+) channels we have co-expressed NCS-1 in Xenopus oocytes, with Ca(V)1.2, Ca(V)2.1, and Ca(V)2.2 Ca(2+) channels, using various subunit combinations. The major effect of NCS-1 was to decrease Ca(2+) current amplitude, recorded with the three different types of alpha(1) subunit. When expressed with Ca(V)2.1, the depression of Ca(2+) current amplitude induced by NCS-1 was dependent upon the identity of the beta subunit expressed, with no block recorded without beta subunit or with the beta(3) subunit. Current-voltage and inactivation curves were also slightly modified and displayed a different specificity toward the beta subunits. Taken together, these data suggest that NCS-1 is able to modulate cardiac and neuronal voltage-gated Ca(2+) channels in a beta subunit specific manner.
Collapse
Affiliation(s)
- Matthieu Rousset
- Centre de Recherche de Biochimie Macromoléculaire, CNRS Unité Propre de Recherche 1086, Institut Federatif de Recherche 24, 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | |
Collapse
|
92
|
Rajebhosale M, Greenwood S, Vidugiriene J, Jeromin A, Hilfiker S. Phosphatidylinositol 4-OH kinase is a downstream target of neuronal calcium sensor-1 in enhancing exocytosis in neuroendocrine cells. J Biol Chem 2003; 278:6075-84. [PMID: 12471042 DOI: 10.1074/jbc.m204702200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal calcium sensor-1 (NCS-1), the mammalian orthologue of frequenin, belongs to a family of EF-hand-containing Ca(2+) sensors. NCS-1/frequenin has been shown to enhance synaptic transmission in PC12 cells and Drosophila and Xenopus, respectively. However, the precise molecular mechanism for the enhancement of exocytosis is largely unknown. In PC12 cells, NCS-1 potentiated exocytosis evoked by ATP, an agonist to phospholipase C-linked receptors, but had no effect on depolarization-evoked release. NCS-1 also enhanced exocytosis triggered by ionomycin, a Ca(2+) ionophore that bypasses K(+) and Ca(2+) channels. Overexpression of NCS-1 caused a shift in the dose-response curve of inhibition of ATP-evoked secretion using phenylarsine oxide, an inhibitor of phosphatidylinositol 4-OH kinase (PI4K). Plasma membrane phosphatidylinositol 4,5-bisphosphate pools were increased upon NCS-1 transfection as visualized using a phospholipase C-delta pleckstrin homology domain-green fluorescent protein construct. NCS-1-transfected cell extracts displayed increased phosphatidylinositol-4-phosphate biosynthesis, indicating an increase in PI4K activity. Mutations in NCS-1 equivalent to those that abolish the interaction of recoverin, another EF-hand-containing Ca(2+) sensor, with its downstream target rhodopsin kinase, lost their ability to enhance exocytosis. Taken together, the present data indicate that NCS-1 modulates the activity of PI4K, leading to increased levels of phosphoinositides and concomitant enhancement of exocytosis.
Collapse
Affiliation(s)
- Manisha Rajebhosale
- University of Manchester, School of Biological Sciences, Oxford Road, Manchester M13 9PT, United Kingdom
| | | | | | | | | |
Collapse
|
93
|
Nadal MS, Ozaita A, Amarillo Y, Vega-Saenz de Miera E, Ma Y, Mo W, Goldberg EM, Misumi Y, Ikehara Y, Neubert TA, Rudy B. The CD26-related dipeptidyl aminopeptidase-like protein DPPX is a critical component of neuronal A-type K+ channels. Neuron 2003; 37:449-61. [PMID: 12575952 DOI: 10.1016/s0896-6273(02)01185-6] [Citation(s) in RCA: 269] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Subthreshold-activating somatodendritic A-type potassium channels have fundamental roles in neuronal signaling and plasticity which depend on their unique cellular localization, voltage dependence, and kinetic properties. Some of the components of A-type K(+) channels have been identified; however, these do not reproduce the properties of the native channels, indicating that key molecular factors have yet to be unveiled. We purified A-type K(+) channel complexes from rat brain membranes and found that DPPX, a protein of unknown function that is structurally related to the dipeptidyl aminopeptidase and cell adhesion protein CD26, is a novel component of A-type K(+) channels. DPPX associates with the channels' pore-forming subunits, facilitates their trafficking and membrane targeting, reconstitutes the properties of the native channels in heterologous expression systems, and is coexpressed with the pore-forming subunits in the somatodendritic compartment of CNS neurons.
Collapse
Affiliation(s)
- Marcela S Nadal
- Department of Physiology and Neuroscience and Department of Biochemistry, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Campos D, Jiménez-Díaz L, Carrión AM. Ca(2+)-dependent prodynorphin transcriptional derepression in neuroblastoma cells is exerted through DREAM protein activity in a kinase-independent manner. Mol Cell Neurosci 2003; 22:135-45. [PMID: 12676525 DOI: 10.1016/s1044-7431(03)00040-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Prodynorphin transcription has been postulated as an important molecular mechanism involved in adaptation/repair processes. Expression of prodynorphin is modulated according to the levels of the second messengers cAMP and Ca(2+). In the neuroblastoma cell lines, the increase of prodynorphin mRNA levels is coupled to an elevation of intracellular cAMP levels. Promoter analyses have revealed that the DRE site, a silencer element present in the prodynorphin promoter, is involved in PKA-dependent prodynorphin derepression. In this way, DREAM, a calcium-dependent repressor, plays an outstanding role. In this study, Ca(2+) release from internal stores has been found to promote an increase of prodynorphin mRNA levels in NB69 cells. Surprisingly, Ca(2+)-dependent prodynorphin gene transcription was insensitive to the broad-spectrum kinase inhibitors and sensitive to agents that alter internal Ca(2+) accumulation. Moreover, we demonstrate that in NB69 cells, the Ca(2+) signaling pathway uses DREAM as an effector to evoke prodynorphin transcription derepression in a kinase-independent manner.
Collapse
Affiliation(s)
- David Campos
- Centro Nacional de Biotecnologia (CNB), Universidad Autónoma de Madrid, Cantoblanco, E-28049, Madrid, Spain
| | | | | |
Collapse
|
95
|
Koh PO, Undie AS, Kabbani N, Levenson R, Goldman-Rakic PS, Lidow MS. Up-regulation of neuronal calcium sensor-1 (NCS-1) in the prefrontal cortex of schizophrenic and bipolar patients. Proc Natl Acad Sci U S A 2003; 100:313-7. [PMID: 12496348 PMCID: PMC140961 DOI: 10.1073/pnas.232693499] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2002] [Indexed: 11/18/2022] Open
Abstract
The delineation of dopamine dysfunction in the mentally ill has been a long-standing quest of biological psychiatry. The present study focuses on a recently recognized group of dopamine receptor-interacting proteins as possible novel sites of dysfunction in schizophrenic and bipolar patients. We demonstrate that the dorsolateral prefrontal cortex in schizophrenia and bipolar cases from the Stanley Foundation Neuropathology Consortium display significantly elevated levels of the D2 dopamine receptor desensitization regulatory protein, neuronal calcium sensor-1. These levels of neuronal calcium sensor-1 were not influenced by age, gender, hemisphere, cause of death, postmortem period, alcohol consumption, or antipsychotic and mood stabilizing medications. The present study supports the hypothesis that schizophrenia and bipolar disorder may be associated with abnormalities in dopamine receptor-interacting proteins.
Collapse
Affiliation(s)
- Phil Ok Koh
- Departments of Oral and Craniofacial Biological Sciences, University of Maryland, Baltimore 21201, USA
| | | | | | | | | | | |
Collapse
|
96
|
Bastianelli E. Distribution of calcium-binding proteins in the cerebellum. CEREBELLUM (LONDON, ENGLAND) 2003; 2:242-62. [PMID: 14964684 DOI: 10.1080/14734220310022289] [Citation(s) in RCA: 201] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Calcium plays a fundamental role in the cell as second messenger and is principally regulated by calcium-binding proteins. Although these proteins share in common their ability to bind calcium, they belong to different subfamilies. They present, in general, specific developmental and distribution patterns. Most Purkinje cells express the fast and slow calcium buffer proteins calbindin-D28k and parvalbumin, whereas basket, stellate and Golgi cells the slow buffer parvalbumin only. They are, almost all, calretinin negative. Granule, Lugaro and unipolar brush cells present an opposite immunoreactivity profile, most of them being calretinin positive while lacking calbindin-D28k and parvalbumin. The developmental pattern of appearance of these proteins seems to follow the maturation of neurons. Calbindin-D28k appears early, shortly after cessation of mitosis when neurons become ready to start migration and differentiation while parvalbumin is expressed later in parallel with an increase in neuronal activity. The other proteins are generally detected later. During development, some of these proteins, like calretinin, are transiently expressed in specific cellular subpopulations. The function of these proteins is not fully understood, although strong evidence supports a prominent role in physiological settings with altered calcium concentrations. These proteins regulate and are regulated by intracellular calcium level. For example, they may directly or indirectly enable sensitization or desensitization of calcium channels, and may further block calcium entry into the cells, like the calcium-sensor proteins, that have been shown to be potent and specific modulators of ion channels, which may allow for feedback control of current function and hence signaling. The absence of calcium buffer proteins results in marked abnormalities in cell firing; with alterations in simple and complex spikes or transformation of depressing synapses into facilitating synapses. Calcium-binding protein implication in resistance to degeneration is still a controversial issue. Neurons rich in calcium-binding proteins, especially calbindin-D28k and parvalbumin, seem to be relatively resistant to degeneration in a variety of acute and chronic disorders. However other data support that an absence of calcium-binding proteins may also have a neuroprotective effect. It is not unlikely that neurons may face a dual action mechanism where a decrease in calcium-binding proteins has a first short-term beneficial effect while it becomes detrimental for the cell over the long term.
Collapse
|
97
|
Ohya S, Horowitz B. Differential transcriptional expression of Ca2+ BP superfamilies in murine gastrointestinal smooth muscles. Am J Physiol Gastrointest Liver Physiol 2002; 283:G1290-7. [PMID: 12388203 DOI: 10.1152/ajpgi.00101.2002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Calmodulin (Cal) plays important roles for contractile activity in smooth muscles. Recently, two distinct Ca(2+)-binding protein superfamilies with sequence similarities to Cal have been identified in neuronal cells: neuronal Ca(2+)-binding proteins (NCBPs) and Cal-like Ca(2+)-binding proteins (CaBPs). Some NCBPs and CaBPs play significant roles for Ca(2+)-dependent cellular signaling in the nervous system. In gastrointestinal smooth muscles (GISMs), Cal functions as the regulator of contractile behavior and electrical rhythmicity. However, the molecular identification of NCBPs and CaBPs has not been elucidated in GISMs. Here, we have identified NCBPs and CaBPs expressed in GISMs and determined the expression levels of their transcripts by quantitative RT-PCR. Of 12 NCBPs, the transcripts for neuronal Ca(2+) sensor 1, neural visinin-like proteins 1, 2, and 3, and K(+) channel-interacting proteins 1 and 3 were detected in proximal colon, gastric fundus, gastric antrum, and jejunum. On the other hand, of seven CaBPs including alternatively spliced variants, only CaBP1L transcripts were detected in GISMs.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557-0046, USA
| | | |
Collapse
|
98
|
Patel SP, Campbell DL, Strauss HC. Elucidating KChIP effects on Kv4.3 inactivation and recovery kinetics with a minimal KChIP2 isoform. J Physiol 2002; 545:5-11. [PMID: 12433945 PMCID: PMC2290650 DOI: 10.1113/jphysiol.2002.031856] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2002] [Accepted: 10/06/2002] [Indexed: 11/08/2022] Open
Abstract
Kv channel interacting proteins (KChIPs) are Ca(2+)-binding proteins with four EF-hands. KChIPs modulate Kv4 channel gating by slowing inactivation kinetics and accelerating recovery kinetics. Thus, KChIPs are believed to be important regulators of Kv4 channels underlying transient outward K(+) currents in many excitable cell types. We have cloned a structurally minimal KChIP2 isoform (KChIP2d) from ferret heart. KChIP2d corresponds to the final 70 C-terminal amino acids of other KChIPs and has only one EF-hand. We demonstrate that KChIP2d is a functional KChIP that both accelerates recovery and slows inactivation kinetics of Kv4.3, indicating that the minimal C-terminus can maintain KChIP regulatory properties. We utilize KChIP2d to further demonstrate that: (i) the EF-hand modulates effects on Kv4.3 inactivation but not recovery; (ii) Ca(2+)-dependent effects on Kv4.3 inactivation are mediated through a mechanism reflected in the slow time constant of inactivation; and (iii) a short stretch of amino acids exclusive of the EF-hand partially mediates Ca(2+)-independent effects on recovery. Our results demonstrate that distinct regions of a KChIP molecule are involved in modulating inactivation and recovery. The potential ability of KChIP EF-hands to sense intracellular Ca(2+) levels and transduce these changes to alterations in Kv4 channel inactivation kinetics may serve as a mechanism allowing intracellular Ca(2+) transients to modulate repolarization. KChIP2d is a valuable tool for elucidating structural domains of KChIPs involved in Kv4 channel regulation.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, 124 Sherman Hall, Buffalo, NY 14214, USA.
| | | | | |
Collapse
|
99
|
Affiliation(s)
- Koichi Takimoto
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | | |
Collapse
|
100
|
Lin L, Jeanclos EM, Treuil M, Braunewell KH, Gundelfinger ED, Anand R. The calcium sensor protein visinin-like protein-1 modulates the surface expression and agonist sensitivity of the alpha 4beta 2 nicotinic acetylcholine receptor. J Biol Chem 2002; 277:41872-8. [PMID: 12202488 DOI: 10.1074/jbc.m206857200] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The calcium sensor protein visinin-like protein-1 (VILIP-1) was isolated from a brain cDNA yeast two-hybrid library using the large cytoplasmic domain of the alpha4 subunit as a bait. VILIP-1 is a myristoylated calcium sensor protein that contains three functional calcium binding EF-hand motifs. The alpha4 subunit residues 302-339 were found to be essential for the interaction with VILIP-1. VILIP-1 coimmunopurified with detergent-solubilized recombinant alpha4beta2 acetylcholine receptors (AChRs) expressed in tsA201 cells and with native alpha4 AChRs isolated from brain. Coexpression of VILIP-1 with recombinant alpha4beta2 AChRs up-regulated their surface expression levels approximately 2-fold and increased their agonist sensitivity to acetylcholine approximately 3-fold. The modulation of the recombinant alpha4beta2 AChRs by VILIP-1 was attenuated in VILIP-1 mutants that lacked the ability to be myristoylated or to bind calcium. Collectively, these results suggest that VILIP-1 represents a novel modulator of alpha4beta2 AChRs that increases their surface expression levels and agonist sensitivity in response to changes in the intracellular levels of calcium.
Collapse
Affiliation(s)
- Lin Lin
- Neuroscience Center of Excellence, Louisiana, New Orleans 70112, USA
| | | | | | | | | | | |
Collapse
|