1
|
Structural mechanisms of gating and selectivity of human rod CNGA1 channel. Neuron 2021; 109:1302-1313.e4. [PMID: 33651975 DOI: 10.1016/j.neuron.2021.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 11/22/2022]
Abstract
Mammalian cyclic nucleotide-gated (CNG) channels play an essential role in the signal transduction of the visual and olfactory sensory systems. Here we reveal the structural mechanism of ligand gating in human rod CNGA1 channel by determining its cryo-EM structures in both the apo closed and cGMP-bound open states. Distinct from most other members of voltage-gated tetrameric cation channels, CNGA1 forms a central channel gate in the middle of the membrane, occluding the central cavity. Structural analyses of ion binding profiles in the selectivity filters of the wild-type channel and the E365Q filter mutant allow us to unambiguously define the two Ca2+ binding sites inside the selectivity filter, providing structural insights into Ca2+ blockage and permeation in CNG channels. The structure of the E365Q mutant also reveals two alternative side-chain conformations at Q365, providing a plausible explanation for the voltage-dependent gating of CNG channel acquired upon E365 mutation.
Collapse
|
2
|
Jarratt-Barnham E, Wang L, Ning Y, Davies JM. The Complex Story of Plant Cyclic Nucleotide-Gated Channels. Int J Mol Sci 2021; 22:ijms22020874. [PMID: 33467208 PMCID: PMC7830781 DOI: 10.3390/ijms22020874] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Plant cyclic nucleotide-gated channels (CNGCs) are tetrameric cation channels which may be activated by the cyclic nucleotides (cNMPs) adenosine 3',5'-cyclic monophosphate (cAMP) and guanosine 3',5'-cyclic monophosphate (cGMP). The genome of Arabidopsis thaliana encodes 20 CNGC subunits associated with aspects of development, stress response and immunity. Recently, it has been demonstrated that CNGC subunits form heterotetrameric complexes which behave differently from the homotetramers produced by their constituent subunits. These findings have widespread implications for future signalling research and may help explain how specificity can be achieved by CNGCs that are known to act in disparate pathways. Regulation of complex formation may involve cyclic nucleotide-gated channel-like proteins.
Collapse
|
3
|
Banerjee S, Yao J, Zhang X, Niu J, Chen Z. Next generation sequencing identified novel heterozygous nonsense mutation in CNGB1 gene associated with retinitis pigmentosa in a Chinese patient. Oncotarget 2017; 8:88345-88350. [PMID: 29179439 PMCID: PMC5687609 DOI: 10.18632/oncotarget.21728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/08/2017] [Indexed: 11/25/2022] Open
Abstract
Retinitis pigmentosa (RP) is a severe hereditary eye disease characterized by progressive degeneration of photoreceptors and subsequent loss of vision. Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous group of retinal diseases. Germline mutations of CNGB1 is associated with retinitis pigmentosa. We have identified and investigated a 34-year-old Chinese man with markedly have night vision blindness and loss of midperipheral visual field. The proband also lose his far peripheral visual field and also central vision. Proband's retinal pigment deposits visible on fundus examination and primary loss of rod photoreceptor cells followed by secondary loss of cone photoreceptors. Target exome capture based next generation sequencing and Sanger sequencing identified novel nonsense mutation, c.1917G>A and a reported mutation, c.2361C>A, in the CNGB1 gene. Both the nonsense mutations are predicted to lead to the formation of a premature stop codon which finally results into formation of truncated CNGB1 protein product which finally predicted to be disease causing. According to the variant classification guidelines of ACMG, these two variants are categorized as "likely pathogenic" variants. Our findings expand the mutational spectra of CNGB1 and are valuable in the mutation-based pre- and post-natal screening and genetic diagnosis for retinitis pigmentosa.
Collapse
Affiliation(s)
- Santasree Banerjee
- Department of Cell Biology and Medical Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junping Yao
- Department of Ophthalmology, Tianyou Hospital Affiliated to Wuhan University of Science & Technology, Wuhan, China
| | - Xinxin Zhang
- Department of Cell Biology and Medical Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianjun Niu
- Department of Ophthalmology, General Hospital of Xinjiang Military Area Command of Chinese PLA, Urumqi, China
| | - Zhongshan Chen
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Ding XQ, Thapa A, Ma H, Xu J, Elliott MH, Rodgers KK, Smith ML, Wang JS, Pittler SJ, Kefalov VJ. The B3 Subunit of the Cone Cyclic Nucleotide-gated Channel Regulates the Light Responses of Cones and Contributes to the Channel Structural Flexibility. J Biol Chem 2016; 291:8721-34. [PMID: 26893377 DOI: 10.1074/jbc.m115.696138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Indexed: 11/06/2022] Open
Abstract
Cone photoreceptor cyclic nucleotide-gated (CNG) channels play a pivotal role in cone phototransduction, which is a process essential for daylight vision, color vision, and visual acuity. Mutations in the cone channel subunits CNGA3 and CNGB3 are associated with human cone diseases, including achromatopsia, cone dystrophies, and early onset macular degeneration. Mutations in CNGB3 alone account for 50% of reported cases of achromatopsia. This work investigated the role of CNGB3 in cone light response and cone channel structural stability. As cones comprise only 2-3% of the total photoreceptor population in the wild-type mouse retina, we used Cngb3(-/-)/Nrl(-/-) mice with CNGB3 deficiency on a cone-dominant background in our study. We found that, in the absence of CNGB3, CNGA3 was able to travel to the outer segments, co-localize with cone opsin, and form tetrameric complexes. Electroretinogram analyses revealed reduced cone light response amplitude/sensitivity and slower response recovery in Cngb3(-/-)/Nrl(-/-) mice compared with Nrl(-/-) mice. Absence of CNGB3 expression altered the adaptation capacity of cones and severely compromised function in bright light. Biochemical analysis demonstrated that CNGA3 channels lacking CNGB3 were more resilient to proteolysis than CNGA3/CNGB3 channels, suggesting a hindered structural flexibility. Thus, CNGB3 regulates cone light response kinetics and the channel structural flexibility. This work advances our understanding of the biochemical and functional role of CNGB3 in cone photoreceptors.
Collapse
Affiliation(s)
| | | | - Hongwei Ma
- From the Departments of Cell Biology and
| | - Jianhua Xu
- From the Departments of Cell Biology and
| | - Michael H Elliott
- Ophthalmology and Dean McGee Eye Institute, Oklahoma City, Oklahoma 73104
| | - Karla K Rodgers
- Biochemistry, University of Oklahoma Health Sciences Center and
| | - Marci L Smith
- Department of Vision Sciences, University of Alabama, Birmingham, Alabama 35924, and
| | - Jin-Shan Wang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Steven J Pittler
- Department of Vision Sciences, University of Alabama, Birmingham, Alabama 35924, and
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri 63110
| |
Collapse
|
5
|
Scholz P, Kalbe B, Jansen F, Altmueller J, Becker C, Mohrhardt J, Schreiner B, Gisselmann G, Hatt H, Osterloh S. Transcriptome Analysis of Murine Olfactory Sensory Neurons during Development Using Single Cell RNA-Seq. Chem Senses 2016; 41:313-23. [PMID: 26839357 DOI: 10.1093/chemse/bjw003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mammalian odor reception is achieved by highly specialized olfactory sensory neurons (OSNs) located in the nasal cavity. Despite their importance for the daily survival of most mammals, the gene expression and regulatory profiles of these single neurons are poorly understood. Here, we report the isolation of individual GFP-labeled OSNs from Olfr73-GFP mice at different developmental stages followed by Next Generation Sequencing, thereby analyzing the detailed transcriptome for the first time. We characterized the repertoire of olfactory receptors (ORs) and found that in addition to the highly and predominant detectable Olfr73, 20 additional ORs were stably detectable at lower transcript levels in adult mice. Additionally, OSNs collected from mice of earlier developmental stages did not show any stable OR patterns. However, more than one predominant OR per OSN was detectable.
Collapse
Affiliation(s)
- Paul Scholz
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Benjamin Kalbe
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Fabian Jansen
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Janine Altmueller
- Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, NRW, Germany and Institute of Human Genetics, University Hospital Cologne, 50931 Cologne, NRW, Germany
| | - Christian Becker
- Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, NRW, Germany and
| | - Julia Mohrhardt
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Benjamin Schreiner
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Guenter Gisselmann
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany
| | - Sabrina Osterloh
- Department of Cell Physiology, Ruhr-University Bochum, 44801 Bochum, NRW, Germany,
| |
Collapse
|
6
|
|
7
|
Hallworth R, Nichols MG. Single molecule imaging approach to membrane protein stoichiometry. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2012; 18:771-780. [PMID: 22831749 PMCID: PMC3786598 DOI: 10.1017/s1431927612001195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Recent technical advances have enabled the imaging of single fluorescent molecules. The application of single molecule visualization techniques has opened up new avenues of experimentation in biology at the molecular level. In this article, we review the application of single fluorescent molecule visualization and analysis to an important problem, that of subunit stoichiometry in membrane proteins, with particular emphasis on our approach. Single fluorescent molecules, coupled to fluorescent proteins, are localized in the membranes of cells. The molecules are then exposed to continuous low-level excitation until their fluorescent emissions reach background levels. The high sensitivity of modern instrumentation has enabled direct observations of discrete step decreases in the fluorescence of single molecules, which represent the bleaching of single fluorophores. By counting the number of steps over a large number of single molecules, an average step count is determined from which the stoichiometry is deduced using a binomial model. We examined the stoichiometry of a protein, prestin, that is central to mammalian hearing. We discuss how we prepared, identified, and imaged single molecules of prestin. The methodological considerations behind our approach are described and compared to similar procedures in other laboratories.
Collapse
Affiliation(s)
- Richard Hallworth
- Department of Biomedical Sciences, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA.
| | | |
Collapse
|
8
|
Nache V, Zimmer T, Wongsamitkul N, Schmauder R, Kusch J, Reinhardt L, Bönigk W, Seifert R, Biskup C, Schwede F, Benndorf K. Differential regulation by cyclic nucleotides of the CNGA4 and CNGB1b subunits in olfactory cyclic nucleotide-gated channels. Sci Signal 2012; 5:ra48. [PMID: 22786723 DOI: 10.1126/scisignal.2003110] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Olfactory cyclic nucleotide-gated (CNG) ion channels are essential contributors to signal transduction of olfactory sensory neurons. The activity of the channels is controlled by the cyclic nucleotides guanosine 3',5'-monophosphate (cGMP) and adenosine 3',5'-monophosphate (cAMP). The olfactory CNG channels are composed of two CNGA2 subunits, one CNGA4 and one CNGB1b subunit, each containing a cyclic nucleotide-binding domain. Using patch-clamp fluorometry, we measured ligand binding and channel activation simultaneously and showed that cGMP activated olfactory CNG channels not only by binding to the two CNGA2 subunits but also by binding to the CNGA4 subunit. In a channel in which the CNGA2 subunits were compromised for ligand binding, cGMP binding to CNGA4 was sufficient to partly activate the channel. In contrast, in heterotetrameric channels, the CNGB1b subunit did not bind cGMP, but channels with this subunit showed activation by cAMP. Thus, the modulatory subunits participate actively in translating ligand binding to activation of heterotetrameric olfactory CNG channels and enable the channels to differentiate between cyclic nucleotides.
Collapse
Affiliation(s)
- Vasilica Nache
- Institute of Physiology II, University Hospital Jena, Friedrich-Schiller-University Jena, D-07740 Jena, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
CNGA3 is expressed in inner ear hair cells and binds to an intracellular C-terminus domain of EMILIN1. Biochem J 2012; 443:463-76. [PMID: 22248097 DOI: 10.1042/bj20111255] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The molecular characteristics of CNG (cyclic nucleotide-gated) channels in auditory/vestibular hair cells are largely unknown, unlike those of CNG mediating sensory transduction in vision and olfaction. In the present study we report the full-length sequence for three CNGA3 variants in a hair cell preparation from the trout saccule with high identity to CNGA3 in olfactory receptor neurons/cone photoreceptors. A custom antibody targeting the N-terminal sequence immunolocalized CNGA3 to the stereocilia and subcuticular plate region of saccular hair cells. The cytoplasmic C-terminus of CNGA3 was found by yeast two-hybrid analysis to bind the C-terminus of EMILIN1 (elastin microfibril interface-located protein 1) in both the vestibular hair cell model and rat organ of Corti. Specific binding between CNGA3 and EMILIN1 was confirmed with surface plasmon resonance analysis, predicting dependence on Ca2+ with Kd=1.6×10-6 M for trout hair cell proteins and Kd=2.7×10-7 M for organ of Corti proteins at 68 μM Ca2+. Pull-down assays indicated that the binding to organ of Corti CNGA3 was attributable to the EMILIN1 intracellular sequence that follows a predicted transmembrane domain in the C-terminus. Saccular hair cells also express the transcript for PDE6C (phosphodiesterase 6C), which in cone photoreceptors regulates the degradation of cGMP used to gate CNGA3 in phototransduction. Taken together, the evidence supports the existence in saccular hair cells of a molecular pathway linking CNGA3, its binding partner EMILIN1 (and β1 integrin) and cGMP-specific PDE6C, which is potentially replicated in cochlear outer hair cells, given stereociliary immunolocalizations of CNGA3, EMILIN1 and PDE6C.
Collapse
|
10
|
Abstract
The unusual membrane motor protein prestin is essential for mammalian hearing and for the survival of cochlear outer hair cells. While prestin has been demonstrated to be a homooligomer, by Western blot and FRET analyses, the stoichiometry of self association is unclear. Prestin, coupled to the enhanced green fluorescent protein, was synthesized and membrane targeted in human embryonic kidney cells by plasmid transfection. Fragments of membrane containing immobilized fluorescent molecules were isolated by osmotic lysis. Diffraction-limited fluorescent spots consistent in size with single molecules were observed. Under continuous excitation, the spots bleached to background in sequential and approximately equal-amplitude steps. The average step count to background levels was 2.7. A binomial model of prestin oligomerization indicated that prestin was most likely a tetramer, and that a fraction of the green fluorescent protein molecules was dark. As a positive control, the same procedure was applied to cells transfected with plasmids coding for the human cyclic nucleotide-gated ion channel A3 subunit (again coupled to the enhanced green fluorescent protein), which is an obligate tetramer. The average step count for this molecule was also 2.7. This result implies that in cell membranes prestin oligomerizes to a tetramer.
Collapse
Affiliation(s)
- Richard Hallworth
- Dept. of Biomedical Sciences, Creighton Univ., 2500 California Plaza, Omaha, NE 68178, USA.
| | | |
Collapse
|
11
|
Moroni M, Meyer JO, Lahmann C, Sivilotti LG. In glycine and GABA(A) channels, different subunits contribute asymmetrically to channel conductance via residues in the extracellular domain. J Biol Chem 2011; 286:13414-22. [PMID: 21343294 DOI: 10.1074/jbc.m110.204610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Single-channel conductance in Cys-loop channels is controlled by the nature of the amino acids in the narrowest parts of the ion conduction pathway, namely the second transmembrane domain (M2) and the intracellular helix. In cationic channels, such as Torpedo ACh nicotinic receptors, conductance is increased by negatively charged residues exposed to the extracellular vestibule. We now show that positively charged residues at the same loop 5 position boost also the conductance of anionic Cys-loop channels, such as glycine (α1 and α1β) and GABA(A) (α1β2γ2) receptors. Charge reversal mutations here produce a greater decrease on outward conductance, but their effect strongly depends on which subunit carries the mutation. In the glycine α1β receptor, replacing Lys with Glu in α1 reduces single-channel conductance by 41%, but has no effect in the β subunit. By expressing concatameric receptors with constrained stoichiometry, we show that this asymmetry is not explained by the subunit copy number. A similar pattern is observed in the α1β2γ2 GABA(A) receptor, where only mutations in α1 or β2 decreased conductance (to different extents). In both glycine and GABA receptors, the effect of mutations in different subunits does not sum linearly: mutations that had no detectable effect in isolation did enhance the effect of mutations carried by other subunits. As in the nicotinic receptor, charged residues in the extracellular vestibule of anionic Cys-loop channels influence elementary conductance. The size of this effect strongly depends on the direction of the ion flow and, unexpectedly, on the nature of the subunit that carries the residue.
Collapse
Affiliation(s)
- Mirko Moroni
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | | | | | | |
Collapse
|
12
|
Hachez C, Chaumont F. Aquaporins: a family of highly regulated multifunctional channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 679:1-17. [PMID: 20666220 DOI: 10.1007/978-1-4419-6315-4_1] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Aquaporins (AQPs) were discovered as channels facilitatingwater movement across cellular membranes. Whereas much of the research has focused on characterizing AQPs with respect to cell water homeostasis, recent discoveries in terms of the transport selectivity of AQP homologs has shed new light on their physiological roles. In fact, whereas some AQPs behave as "strict" water channels, others can conduct a wide range ofnonpolar solutes, such as urea or glycerol and even more unconventional permeants, such as the nonpolar gases carbon dioxide and nitric oxide, the polar gas ammonia, the reactive oxygen species hydrogen peroxide and the metalloids antimonite, arsenite, boron and silicon. This suggests that AQPs are also key players in various physiological processes not related to water homeostasis. The function, regulation and biological importance of AQPs in the different kingdoms is reviewed in this chapter, with special emphasis on animal and plant AQPs.
Collapse
Affiliation(s)
- Charles Hachez
- Institut des Sciences de la Vie, Universit4 catholique de Louvain, Croix du Sud 5-15, B-1348 Louvain-la-Neuve, Belgium
| | | |
Collapse
|
13
|
Lebaudy A, Pascaud F, Véry AA, Alcon C, Dreyer I, Thibaud JB, Lacombe B. Preferential KAT1-KAT2 heteromerization determines inward K+ current properties in Arabidopsis guard cells. J Biol Chem 2009; 285:6265-74. [PMID: 20040603 DOI: 10.1074/jbc.m109.068445] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Guard cells adjust their volume by changing their ion content due to intense fluxes that, for K(+), are believed to flow through inward or outward Shaker channels. Because Shaker channels can be homo- or heterotetramers and Arabidopsis guard cells express at least five genes encoding inward Shaker subunits, including the two major ones, KAT1 and KAT2, the molecular identity of inward Shaker channels operating therein is not yet completely elucidated. Here, we first addressed the properties of KAT1-KAT2 heteromers by expressing KAT1-KAT2 tandems in Xenopus oocytes. Then, computer analyses of the data suggested that coexpression of free KAT1 and KAT2 subunits resulted mainly in heteromeric channels made of two subunits of each type due to some preferential association of KAT1-KAT2 heterodimers at the first step of channel assembly. This was further supported by the analysis of KAT2 effect on KAT1 targeting in tobacco cells. Finally, patch-clamp recordings of native inward channels in wild-type and mutant genotypes strongly suggested that this preferential heteromerization occurs in planta and that Arabidopsis guard cell inward Shaker channels are mainly heteromers of KAT1 and KAT2 subunits.
Collapse
Affiliation(s)
- Anne Lebaudy
- Biochimie et Physiologie Moléculaire des Plantes, CNRS UMR 5004, Institut National de la Recherche Agronomique U386, Montpellier SupAgro, Université Montpellier II, Place Viala, 34060 Montpellier Cedex, France
| | | | | | | | | | | | | |
Collapse
|
14
|
Wang Z, Jiang Y, Lu L, Huang R, Hou Q, Shi F. Molecular mechanisms of cyclic nucleotide-gated ion channel gating. J Genet Genomics 2009; 34:477-85. [PMID: 17601606 DOI: 10.1016/s1673-8527(07)60052-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Accepted: 01/08/2007] [Indexed: 01/09/2023]
Abstract
Cyclic nucleotide-gated ion channels (CNGs) are distributed most widely in the neuronal cell. Great progress has been made in molecular mechanisms of CNG channel gating in the recent years. Results of many experiments have indicated that the stoichiometry and assembly of CNG channels affect their property and gating. Experiments of CNG mutants and analyses of cysteine accessibilities show that cyclic nucleotide-binding domains (CNBD) bind cyclic nucleotides and subsequently conformational changes occurred followed by the concerted or cooperative conformational change of all four subunits during CNG gating. In order to provide theoretical assistances for further investigation on CNG channels, especially regarding the disease pathogenesis of ion channels, this paper reviews the latest progress on mechanisms of CNG channels, functions of subunits, processes of subunit assembly, and conformational changes of subunit regions during gating.
Collapse
Affiliation(s)
- Zhengchao Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | | | |
Collapse
|
15
|
Zhang P, Luo Y, Chasan B, González-Perrett S, Montalbetti N, Timpanaro GA, Cantero MDR, Ramos AJ, Goldmann WH, Zhou J, Cantiello HF. The multimeric structure of polycystin-2 (TRPP2): structural-functional correlates of homo- and hetero-multimers with TRPC1. Hum Mol Genet 2009; 18:1238-51. [PMID: 19193631 DOI: 10.1093/hmg/ddp024] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Polycystin-2 (PC2, TRPP2), the gene product of PKD2, whose mutations cause autosomal dominant polycystic kidney disease (ADPKD), belongs to the superfamily of TRP channels. PC2 is a non-selective cation channel, with multiple subconductance states. In this report, we explored structural and functional properties of PC2 and whether the conductance substates represent monomeric contributions to the channel complex. A kinetic analysis of spontaneous channel currents of PC2 showed that four intrinsic, non-stochastic subconductance states, which followed a staircase behavior, were both pH- and voltage-dependent. To confirm the oligomeric contributions to PC2 channel function, heteromeric PC2/TRPC1 channel complexes were also functionally assessed by single channel current analysis. Low pH inhibited the PC2 currents in PC2 homomeric complexes, but failed to affect PC2 currents in PC2/TRPC1 heteromeric complexes. Amiloride, in contrast, abolished PC2 currents in both the homomeric PC2 complexes and the heteromeric PC2/TRPC1 complexes, thus PC2/TRPC1 complexes have distinct functional properties from the homomeric complexes. The topological features of the homomeric PC2-, TRPC1- and heteromeric PC2/TRPC1 channel complexes, assessed by atomic force microscopy, were consistent with structural tetramers. TRPC1 homomeric channels had different average diameter and protruding height when compared with the PC2 homomers. The contribution of individual monomers to the PC2/TRPC1 hetero-complexes was easily distinguishable. The data support tetrameric models of both the PC2 and TRPC1 channels, where the overall conductance of a particular channel will depend on the contribution of the various functional monomers in the complex.
Collapse
Affiliation(s)
- Peng Zhang
- Nephrology Division and Electrophysiology Core, Massachusetts General Hospital East, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Sack JT, Shamotienko O, Dolly JO. How to validate a heteromeric ion channel drug target: assessing proper expression of concatenated subunits. ACTA ACUST UNITED AC 2008; 131:415-20. [PMID: 18411330 PMCID: PMC2346572 DOI: 10.1085/jgp.200709939] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jon T Sack
- ternational Centre for Neurotherapeutics, Dublin City University, Dublin 9, Ireland.
| | | | | |
Collapse
|
17
|
Shoshan-Barmatz V, Zakar M, Shmuelivich F, Nahon E, Vardi N. Retina expresses a novel variant of the ryanodine receptor. Eur J Neurosci 2007; 26:3113-25. [PMID: 18005065 DOI: 10.1111/j.1460-9568.2007.05931.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Calcium released from intracellular stores via the ryanodine receptor (RyR) mediates a variety of signalling processes. We previously showed that retina expresses the three known types of RyR, but retinal membrane preparations exhibit unique characteristics such as Ca2+-independent [3H]ryanodine-binding and inhibition by caffeine. We have heretofore suggested that the major retinal RyR isoform is novel. The present study aimed to identify this receptor isoform and to localize RyR in mammalian retina. Immunoblotting with specific and pan-antibodies showed that the major retinal RyR has a mobility similar to that of RyR2 or RyR3. Real-time PCR revealed that the major type is RyR2, and RT-PCR followed by sequencing showed a transcript that encodes a protein with approximately 99% identity to RyR2, yet lacking two regions of seven and 12 amino acids and including an additional insertion of eight amino acids. An antibody against RyR2 localized this type to somas and primary dendrites of most retinal neurons. An antibody against RyR1 localized RyR to most somas but also revealed staining in photoreceptor outer segments, concentrated on the disk membranes at their rim. The ryanodine-binding properties and the electrophoretic mobility of RyR from the outer segments were similar to those of the whole retinal preparation. The results thus identify a novel variant of RyR2 which can contribute to regulating photoreceptor Ca2+ concentrations. The restricted localization of the outer segment RyR to the disk rim suggests that its activation mechanism involves a coupling between retinal RyR and the cGMP-gated channel.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben Gurion University of the Negev, Beer Sheva, 84105, Israel.
| | | | | | | | | |
Collapse
|
18
|
Anantharam A, Palmer LG. Determination of epithelial Na+ channel subunit stoichiometry from single-channel conductances. ACTA ACUST UNITED AC 2007; 130:55-70. [PMID: 17562820 PMCID: PMC2154365 DOI: 10.1085/jgp.200609716] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The epithelial Na+ channel (ENaC) is a multimeric membrane protein consisting of three subunits, α, β, and γ. The total number of subunits per functional channel complex has been described variously to follow either a tetrameric arrangement of 2α:1β:1γ or a higher-ordered stoichiometry of 3α:3β:3γ. Therefore, while it is clear that all three ENaC subunits are required for full channel activity, the number of the subunits required remains controversial. We used a new approach, based on single-channel measurements in Xenopus oocytes to address this issue. Individual mutations that alter single-channel conductance were made in pore-lining residues of ENaC α, β, or γ subunits. Recordings from patches in oocytes expressing a single species, wild type or mutant, of α, β, and γ showed a well-defined current transition amplitude with a single Gaussian distribution. When cRNAs for all three wild-type subunits were mixed with an equimolar amount of a mutant α-subunit (either S589D or S592T), amplitudes corresponding to pure wild-type or mutant conductances could be observed in the same patch, along with a third intermediate amplitude most likely arising from channels with at least one wild-type and at least 1 mutant α-subunit. However, intermediate or hybrid conductances were not observed with coexpression of wild-type and mutant βG529A or γG534E subunits. Our results support a tetrameric arrangement of ENaC subunits where 2α, 1β, and 1γ come together around central pore.
Collapse
Affiliation(s)
- Arun Anantharam
- Graduate Program in Neuroscience, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | |
Collapse
|
19
|
Xicluna J, Lacombe B, Dreyer I, Alcon C, Jeanguenin L, Sentenac H, Thibaud JB, Chérel I. Increased functional diversity of plant K+ channels by preferential heteromerization of the shaker-like subunits AKT2 and KAT2. J Biol Chem 2007; 282:486-94. [PMID: 17085433 DOI: 10.1074/jbc.m607607200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Assembly of plant Shaker subunits as heterotetramers, increasing channel functional diversity, has been reported. Here we focus on a new interaction, between AKT2 and KAT2 subunits. The assembly as AKT2/KAT2 heterotetramers is demonstrated by (i) a strong signal in two-hybrid tests with intracytoplasmic C-terminal regions, (ii) the effect of KAT2 on AKT2 subunit targeting in tobacco cells, (iii) the complete inhibition of AKT2 currents by co-expression with a dominant-negative KAT2 subunit in Xenopus oocytes, and reciprocally, and (iv) the appearance, upon co-expression of wild-type AKT2 and KAT2 subunits, of new channel functional properties that cannot be explained by the co-existence of two kinds of homotetrameric channels. In particular, the instantaneous current, characteristic of AKT2, displayed new functional features when compared with those of AKT2 homotetramers: activation by external acidification (instead of inhibition) and weak inhibition by calcium. Single channel current measurements in oocytes co-expressing AKT2 and KAT2 revealed a strong preference for incorporation of subunits into heteromultimers and a diversity of individual channels. In planta, these new channels, which may undergo specific regulations, are likely to be formed in guard cells and in the phloem, where they could participate in the control of membrane potential and potassium fluxes.
Collapse
Affiliation(s)
- Jérôme Xicluna
- Laboratoire de Biochimie et Physiologie Moléculaire des Plantes, UMR 5004 Agro-Montpellier/Centre National de la Recherche Scientifique/Institut National de la Recherche Agronomique/Université Montpellier II, Place Viala, 34060 Montpellier Cedex 1, France
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Naso A, Montisci R, Gambale F, Picco C. Stoichiometry studies reveal functional properties of KDC1 in plant shaker potassium channels. Biophys J 2006; 91:3673-83. [PMID: 16920836 PMCID: PMC1630452 DOI: 10.1529/biophysj.106.091777] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Functional heteromeric plant Shaker potassium channels can be formed by the assembly of subunits from different tissues, as well as from diverse plant species. KDC1 (K(+) Daucus carota 1) produces inward-rectifying currents in Xenopus oocytes when coexpressed with KAT1 and other subunits appertaining to different plant Shaker subfamilies. Owing to the presence of KDC1, resulting heteromeric channels display slower activation kinetics, a shift of the activation threshold toward more negative membrane potentials and current potentiation upon the addition of external zinc. Despite available information on heteromerization of plant Shaker channels, very little is known to date on the properties of the various stoichiometric configurations formed by different subunits. To investigate the functional properties of heteromeric nKDC1/mKAT1 configurations, we realized a series of dimeric constructs combining KDC1 and KAT1 alpha-subunits. We found that homomeric channels, formed by monomeric or dimeric alpha-subunit constructs, show identical biophysical characteristics. Coinjections of diverse tandem constructs, instead, displayed significantly different currents proving that KDC1 has high affinity for KAT1 and participates in the formation of functional channels with at most two KDC1 subunits, whereas three KDC1 subunits prevented the formation of functional channels. This article brings a contribution to the understanding of the molecular mechanisms regulating plant Shaker channel functionality by association of modulatory subunits.
Collapse
Affiliation(s)
- Alessia Naso
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genoa, Italy
| | | | | | | |
Collapse
|
21
|
Li M, Jiang J, Yue L. Functional characterization of homo- and heteromeric channel kinases TRPM6 and TRPM7. ACTA ACUST UNITED AC 2006; 127:525-37. [PMID: 16636202 PMCID: PMC2151519 DOI: 10.1085/jgp.200609502] [Citation(s) in RCA: 291] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
TRPM6 and TRPM7 are two known channel kinases that play important roles in various physiological processes, including Mg2+ homeostasis. Mutations in TRPM6 cause hereditary hypomagnesemia and secondary hypocalcemia (HSH). However, whether TRPM6 encodes functional channels is controversial. Here we demonstrate several signature features of TRPM6 that distinguish TRPM6 from TRPM7 and TRPM6/7 channels. We show that heterologous expression of TRPM6 but not the mutant TRPM6S141L produces functional channels with divalent cation permeability profile and pH sensitivity distinctive from those of TRPM7 channels and TRPM6/7 complexes. TRPM6 exhibits unique unitary conductance that is 2- and 1.5-fold bigger than that of TRPM7 and TRPM6/7. Moreover, micromolar levels of 2-aminoethoxydiphenyl borate (2-APB) maximally increase TRPM6 but significantly inhibit TRPM7 channel activities; whereas millimolar concentrations of 2-APB potentiate TRPM6/7 and TRPM7 channel activities. Furthermore, Mg2+ and Ca2+ entry through TRPM6 is enhanced three- to fourfold by 2-APB. Collectively, these results indicate that TRPM6 forms functional homomeric channels as well as heteromeric TRPM6/7 complexes. The unique characteristics of these three channel types, TRPM6, TRPM7, and TRPM6/7, suggest that they may play different roles in vivo.
Collapse
Affiliation(s)
- Mingjiang Li
- Center for Cardiology and Cardiovascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington 06030, USA
| | | | | |
Collapse
|
22
|
Contreras JE, Holmgren M. Access of quaternary ammonium blockers to the internal pore of cyclic nucleotide-gated channels: implications for the location of the gate. ACTA ACUST UNITED AC 2006; 127:481-94. [PMID: 16606688 PMCID: PMC2151523 DOI: 10.1085/jgp.200509440] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cyclic nucleotide-gated (CNG) channels play important roles in the transduction of visual and olfactory information by sensing changes in the intracellular concentration of cyclic nucleotides. We have investigated the interactions between intracellularly applied quaternary ammonium (QA) ions and the alpha subunit of rod cyclic nucleotide-gated channels. We have used a family of alkyl-triethylammonium derivatives in which the length of one chain is altered. These QA derivatives blocked the permeation pathway of CNG channels in a concentration- and voltage-dependent manner. For QA compounds with tails longer than six methylene groups, increasing the length of the chain resulted in higher apparent affinities of approximately 1.2 RT per methylene group added, which is consistent with the presence of a hydrophobic pocket within the intracellular mouth of the channel that serves as part of the receptor binding site. At the single channel level, decyltriethyl ammonium (C10-TEA) ions did not change the unitary conductance but they did reduce the apparent mean open time, suggesting that the blocker binds to open channels. We provide four lines of evidence suggesting that QA ions can also bind to closed channels: (1) the extent of C10-TEA blockade at subsaturating [cGMP] was larger than at saturating agonist concentration, (2) under saturating concentrations of cGMP, cIMP, or cAMP, blockade levels were inversely correlated with the maximal probability of opening achieved by each agonist, (3) in the closed state, MTS reagents of comparable sizes to QA ions were able to modify V391C in the inner vestibule of the channel, and (4) in the closed state, C10-TEA was able to slow the Cd2+ inhibition observed in V391C channels. These results are in stark contrast to the well-established QA blockade mechanism in Kv channels, where these compounds can only access the inner vestibule in the open state because the gate that opens and closes the channel is located cytoplasmically with respect to the binding site of QA ions. Therefore, in the context of Kv channels, our observations suggest that the regions involved in opening and closing the permeation pathways in these two types of channels are different.
Collapse
Affiliation(s)
- Jorge E Contreras
- Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
| | | |
Collapse
|
23
|
Brown RL, Strassmaier T, Brady JD, Karpen JW. The pharmacology of cyclic nucleotide-gated channels: emerging from the darkness. Curr Pharm Des 2006; 12:3597-613. [PMID: 17073662 PMCID: PMC2467446 DOI: 10.2174/138161206778522100] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclic nucleotide-gated (CNG) ion channels play a central role in vision and olfaction, generating the electrical responses to light in photoreceptors and to odorants in olfactory receptors. These channels have been detected in many other tissues where their functions are largely unclear. The use of gene knockouts and other methods have yielded some information, but there is a pressing need for potent and specific pharmacological agents directed at CNG channels. To date there has been very little systematic effort in this direction - most of what can be termed CNG channel pharmacology arose from testing reagents known to target protein kinases or other ion channels, or by accident when researchers were investigating other intracellular pathways that may regulate the activity of CNG channels. Predictably, these studies have not produced selective agents. However, taking advantage of emerging structural information and the increasing knowledge of the biophysical properties of these channels, some promising compounds and strategies have begun to emerge. In this review we discuss progress on two fronts, cyclic nucleotide analogs as both activators and competitive inhibitors, and inhibitors that target the pore or gating machinery of the channel. We also discuss the potential of these compounds for treating certain forms of retinal degeneration.
Collapse
Affiliation(s)
- R. Lane Brown
- Neurological Sciences Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Timothy Strassmaier
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - James D. Brady
- Neurological Sciences Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeffrey W. Karpen
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
24
|
Strassmaier T, Uma R, Ghatpande AS, Bandyopadhyay T, Schaffer M, Witte J, McDougal PG, Brown RL, Karpen JW. Modifications to the tetracaine scaffold produce cyclic nucleotide-gated channel blockers with widely varying efficacies. J Med Chem 2005; 48:5805-12. [PMID: 16134947 PMCID: PMC2467444 DOI: 10.1021/jm0502485] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Five new tetracaine analogues were synthesized and evaluated for potency of blockade of cyclic nucleotide-gated channels relative to a multiply charged tetracaine analogue described previously. Increased positive charge at the tertiary amine end of tetracaine results in higher potency and voltage dependence of block. Modifications that reduce the hydrophobic character at the butyl tail are deleterious to block. The tetracaine analogues described here have apparent affinities for CNGA1 channels that vary over nearly 8 orders of magnitude.
Collapse
Affiliation(s)
- Timothy Strassmaier
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Ramalinga Uma
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Ambarish S. Ghatpande
- Department of Physiology and Biophysics, University of Colorado Health Sciences Center, Denver, Colorado 80262
| | - Tapasree Bandyopadhyay
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | | | - John Witte
- Department of Chemistry, Reed College, Portland, Oregon 97202
| | | | - R. Lane Brown
- Neurological Sciences Institute, Oregon Health & Science University, Portland, Oregon 97239
| | - Jeffrey W. Karpen
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
- * Corresponding author: Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239. Phone 503-494-7463; e-mail
| |
Collapse
|
25
|
Abstract
Stimulus-secretion coupling is an essential process in secretory cells in which regulated exocytosis occurs, including neuronal, neuroendocrine, endocrine, and exocrine cells. While an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) is the principal signal, other intracellular signals also are important in regulated exocytosis. In particular, the cAMP signaling system is well known to regulate and modulate exocytosis in a variety of secretory cells. Until recently, it was generally thought that the effects of cAMP in regulated exocytosis are mediated by activation of cAMP-dependent protein kinase (PKA), a major cAMP target, followed by phosphorylation of the relevant proteins. Although the involvement of PKA-independent mechanisms has been suggested in cAMP-regulated exocytosis by pharmacological approaches, the molecular mechanisms are unknown. Newly discovered cAMP-GEF/Epac, which belongs to the cAMP-binding protein family, exhibits guanine nucleotide exchange factor activities and exerts diverse effects on cellular functions including hormone/transmitter secretion, cell adhesion, and intracellular Ca(2+) mobilization. cAMP-GEF/Epac mediates the PKA-independent effects on cAMP-regulated exocytosis. Thus cAMP regulates and modulates exocytosis by coordinating both PKA-dependent and PKA-independent mechanisms. Localization of cAMP within intracellular compartments (cAMP compartmentation or compartmentalization) may be a key mechanism underlying the distinct effects of cAMP in different domains of the cell.
Collapse
Affiliation(s)
- Susumu Seino
- Division of Cellular and Molecular Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | | |
Collapse
|
26
|
Mio K, Ogura T, Hara Y, Mori Y, Sato C. The non-selective cation-permeable channel TRPC3 is a tetrahedron with a cap on the large cytoplasmic end. Biochem Biophys Res Commun 2005; 333:768-77. [PMID: 15964551 DOI: 10.1016/j.bbrc.2005.05.181] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Accepted: 05/31/2005] [Indexed: 10/25/2022]
Abstract
TRPC3 plays important roles in neuronal differentiation and immune cell maturation by mediating the cationic current in response to phospholipase C activation, Ca2+ depletion, and diacylglycerol stimulation. Here, we purified the TRPC3 channel using a glycosylated tetramer and observed the structure using electron microscopy. Negatively stained specimens demonstrate homogeneous protein particles containing an internal cavity-like structure. These particle images were picked up by automated pick-up programs, aligned, and classified by the growing neural gas network method. Similarly oriented projections were averaged to decrease the signal-to-noise ratio. The averaged images progress from the top view to the side views, which are representative of their raw images. The top view confirmed the hypothesis of a four-domain structure, and the side view demonstrates a large cytoplasmic domain with a capped structure at the bottom, which is near a predicted locus of ion release. The total image of the protein is a blunt-edged trapezoid of 200 x 200 x 235 A. This large dimension of TRPC3 is also supported by the Stokes radius (92 A) obtained from gel filtration chromatography.
Collapse
Affiliation(s)
- Kazuhiro Mio
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Umezono 1-1-4, Tsukuba, Ibaraki 305-8568, Japan
| | | | | | | | | |
Collapse
|
27
|
Liu C, Varnum MD. Functional consequences of progressive cone dystrophy-associated mutations in the human cone photoreceptor cyclic nucleotide-gated channel CNGA3 subunit. Am J Physiol Cell Physiol 2005; 289:C187-98. [PMID: 15743887 DOI: 10.1152/ajpcell.00490.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Progressive cone dystrophies are a genetically heterogeneous group of disorders characterized by early deterioration of visual acuity and color vision, together with psychophysical and electrophysiological evidence of abnormal cone function and cone degeneration. Recently, three mutations in the gene encoding the CNGA3 subunit of cone photoreceptor cyclic nucleotide-gated (CNG) channels have been linked to progressive cone dystrophy in humans. To investigate the functional consequences of these mutations, we expressed mutant human CNGA3 subunits in Xenopus oocytes, alone or together with human CNGB3, and studied these channels using patch-clamp recording. Compared with wild-type channels, homomeric and heteromeric channels containing CNGA3-N471S or CNGA3-R563H subunits exhibited an increase in apparent affinity for cGMP and an increase in the relative agonist efficacy of cAMP compared with cGMP. In contrast, R277C subunits did not form functional homomeric or heteromeric channels. Cell surface expression levels, determined using confocal microscopy of green fluorescent protein-tagged subunits and patch-clamp recording, were significantly reduced for both R563H and R277C but unchanged for N471S. Overall, these results suggest that the plasma membrane localization and gating properties of cone CNG channels are altered by progressive cone dystrophy-associated mutations, providing evidence that supports the pathogenicity of these mutations.
Collapse
Affiliation(s)
- Chunming Liu
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology and Program in Neuroscience, Washington State University, PO Box 646520, Pullman, Washington 99164-6520, USA
| | | |
Collapse
|
28
|
Hellwig N, Albrecht N, Harteneck C, Schultz G, Schaefer M. Homo- and heteromeric assembly of TRPV channel subunits. J Cell Sci 2005; 118:917-28. [PMID: 15713749 DOI: 10.1242/jcs.01675] [Citation(s) in RCA: 232] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The vanilloid receptor-related TRP channels (TRPV1-6) mediate thermosensation, pain perception and epithelial Ca(2+) entry. As the specificity of TRPV channel heteromerization and determinants governing the assembly of TRPV subunits were largely elusive, we investigated the TRPV homo- and heteromultimerization. To analyze the assembly of TRPV subunits in living cells, we generated fluorescent fusion proteins or FLAG-tagged TRPV channel subunits. The interaction between TRPV subunits was assessed by analysis of the subcellular colocalization, fluorescence resonance energy transfer and coimmunoprecipitation. Our results demonstrate that TRPV channel subunits do not combine arbitrarily. With the exception of TRPV5 and TRPV6, TRPV channel subunits preferentially assemble into homomeric complexes. Truncation of TRPV1, expression of cytosolic termini of TRPV1 or TRPV4 and construction of chimeric TRPV channel subunits revealed that the specificity and the affinity of the subunit interaction is synergistically provided by interaction modules located in the transmembrane domains and in the cytosolic termini. The relative contribution of intramolecularly linked interaction modules presumably controls the overall affinity and the specificity of TRPV channel assembly.
Collapse
Affiliation(s)
- Nicole Hellwig
- Institut für Pharmakologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Thielallee 67-73, 14195 Berlin, Germany
| | | | | | | | | |
Collapse
|
29
|
Zhou L, Olivier NB, Yao H, Young EC, Siegelbaum SA. A conserved tripeptide in CNG and HCN channels regulates ligand gating by controlling C-terminal oligomerization. Neuron 2005; 44:823-34. [PMID: 15572113 DOI: 10.1016/j.neuron.2004.11.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 09/28/2004] [Accepted: 11/03/2004] [Indexed: 12/17/2022]
Abstract
Cyclic nucleotides directly enhance the opening of the tetrameric CNG and HCN channels, although the mechanism remains unclear. We examined why HCN and certain CNG subunits form functional homomeric channels, whereas other CNG subunits only function in heteromeric channels. The "defect" in the CNGA4 subunit that prevents its homomeric expression was localized to its C-linker, which connects the transmembrane domain to the binding domain and contains a tripeptide that decreases the efficacy of ligand gating. Remarkably, replacement of the homologous HCN tripeptide with the CNGA4 sequence transformed cAMP into an inverse agonist that inhibits HCN channel opening. Using analytical ultracentrifugation, we identified the structural basis for this gating switch: whereas cAMP normally enhances the assembly of HCN C-terminal domains into a tetrameric gating ring, inclusion of the CNGA4 tripeptide reversed this action so that cAMP now causes gating ring disassembly. Thus, ligand gating depends on the dynamic oligomerization of C-terminal binding domains.
Collapse
Affiliation(s)
- Lei Zhou
- Center for Neurobiology and Behavior, Columbia University, 722 West 168 Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
30
|
Zheng J, Zagotta WN. Stoichiometry and assembly of olfactory cyclic nucleotide-gated channels. Neuron 2004; 42:411-21. [PMID: 15134638 DOI: 10.1016/s0896-6273(04)00253-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2004] [Revised: 04/12/2004] [Accepted: 04/14/2004] [Indexed: 11/29/2022]
Abstract
Native ion channels are precisely tuned to their physiological role in neuronal signaling. This tuning frequently involves the controlled assembly of heteromeric channels comprising multiple types of subunits. Cyclic nucleotide-gated (CNG) channels of olfactory neurons are tetramers and require three types of subunits, CNGA2, CNGA4, and CNGB1b, to exhibit properties necessary for olfactory transduction. Using fluorescently tagged subunits and fluorescence resonance energy transfer (FRET), we find the subunit composition of heteromeric olfactory channels in the surface membrane is fixed, with 2:1:1 CNGA2:CNGA4:CNGB1b. Furthermore, when expressed individually with CNGA2, CNGA4 and CNGB1b subunits were still present in only a single copy and, when expressed alone, did not self-assemble. These results suggest that the precise assembly of heteromeric olfactory channels results from a mechanism where CNGA4 and CNGB1b subunits have a high affinity for CNGA2 but not for self-assembly, precluding more than one CNGA4 or CNGB1b subunit in the channel complex.
Collapse
Affiliation(s)
- Jie Zheng
- Howard Hughes Medical Institute, Department of Physiology and Biophysics, Box 357290, University of Washington School of Medicine, Seattle, WA 98195 USA
| | | |
Collapse
|
31
|
Peng C, Rich ED, Varnum MD. Subunit configuration of heteromeric cone cyclic nucleotide-gated channels. Neuron 2004; 42:401-10. [PMID: 15134637 DOI: 10.1016/s0896-6273(04)00225-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2003] [Revised: 01/12/2004] [Accepted: 04/02/2004] [Indexed: 10/25/2022]
Abstract
Cone photoreceptor cyclic nucleotide-gated (CNG) channels are thought to be tetrameric assemblies of CNGB3 (B3) and CNGA3 (A3) subunits. We have used functional and biochemical approaches to investigate the stoichiometry and arrangement of these subunits in recombinant channels. First, tandem dimers of linked subunits were used to constrain the order of CNGB3 and CNGA3 subunits; the properties of channels formed by B3/B3+A3/A3 dimers, or A3/B3+B3/A3 dimers, closely resembled those of channels arising from B3+A3 monomers. Functional markers in B3/B3 (or A3/A3) dimers confirmed that both B3 subunits (and both A3 subunits) gained membership into the pore-forming tetramer and that like subunits were positioned adjacent to each other. Second, chemical crosslinking and co-immunoprecipitation studies using epitope-tagged monomer subunits both demonstrated the presence of two CNGB3 subunits in cone channels. Together, these data support a preferred subunit arrangement for cone CNG channels (B3-B3-A3-A3) that is distinct from the 3A:1B configuration of rod channels.
Collapse
Affiliation(s)
- Changhong Peng
- Department of Veterinary and Comparative Anatomy, Washington State University, P.O. Box 646520, Pullman, WA 99164, USA
| | | | | |
Collapse
|
32
|
Abstract
Cyclic nucleotide-gated (CNG) ion channels were first discovered in rod photoreceptors, where they are responsible for the primary electrical signal of the photoreceptor in response to light. CNG channels are highly specialized membrane proteins that open an ion-permeable pore across the membrane in response to the direct binding of intracellular cyclic nucleotides. CNG channels have been identified in a number of other tissues, including the brain, where their roles are only beginning to be appreciated. Recently, significant progress has been made in understanding the molecular mechanisms underlying their functional specializations. From these studies, a picture is beginning to emerge for how the binding of cyclic nucleotide is transduced into the opening of the pore and how this allosteric transition is modulated by various physiological effectors.
Collapse
Affiliation(s)
- Kimberly Matulef
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA.
| | | |
Collapse
|
33
|
Chérel I. Regulation of K+ channel activities in plants: from physiological to molecular aspects. JOURNAL OF EXPERIMENTAL BOTANY 2004; 55:337-51. [PMID: 14739260 DOI: 10.1093/jxb/erh028] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Plant voltage-gated channels belonging to the Shaker family participate in sustained K+ transport processes at the cell and whole plant levels, such as K+ uptake from the soil solution, long-distance K+ transport in the xylem and phloem, and K+ fluxes in guard cells during stomatal movements. The attention here is focused on the regulation of these transport systems by protein-protein interactions. Clues to the identity of the regulatory mechanisms have been provided by electrophysiological approaches in planta or in heterologous systems, and through analogies with their animal counterparts. It has been shown that, like their animal homologues, plant voltage-gated channels can assemble as homo- or heterotetramers associating polypeptides encoded by different Shaker genes, and that they can bind auxiliary subunits homologous to those identified in mammals. Furthermore, several regulatory processes (involving, for example, protein kinases and phosphatases, G proteins, 14-3-3s, or syntaxins) might be common to plant and animal Shakers. However, the molecular identification of plant channel partners is still at its beginning. This paper reviews current knowledge on plant K+ channel regulation at the physiological and molecular levels, in the light of the corresponding knowledge in animal cells, and discusses perspectives for the deciphering of regulatory networks in the future.
Collapse
Affiliation(s)
- Isabelle Chérel
- Biochimie et Physiologie Moléculaire des Plantes, UMR 5004, Agro-M/INRA/CNRS/UM2, Montpellier, France.
| |
Collapse
|
34
|
Young EC, Krougliak N. Distinct structural determinants of efficacy and sensitivity in the ligand-binding domain of cyclic nucleotide-gated channels. J Biol Chem 2003; 279:3553-62. [PMID: 14594805 DOI: 10.1074/jbc.m310545200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic nucleotide-gated (CNG) channels open in response to direct binding of cyclic nucleotide messengers. Every subunit in a tetrameric CNG channel contains a cytoplasmic ligand-binding domain (BD) that includes a beta-roll (flanked by short helices) and a single C-terminal helix called the C-helix that was previously found to control efficacy (maximal open probability) and selectivity for cGMP versus cAMP. We constructed a series of chimeric CNG channel subunits, each containing a distinct BD sequence (chosen from among six phylogenetically divergent isoforms) fused to an invariant non-BD sequence. We assayed these "BD substitution" chimeras as homomeric CNG channels in Xenopus oo-cytes to compare their functions and found that the most efficient activation by both cAMP and cGMP derived from the BD of the catfish CNGA4 olfactory modulatory subunit (fCNGA4). We then tested the effects of replacing subregions of the bovine CNGA1 BD with corresponding fCNGA4 sequence and hence identified parts of the fCNGA4 BD producing efficient activation. For instance, replacing either the "hinge" that connects the roll and C-helix subdomains or the BD sequence N-terminal to the hinge greatly enhanced cAMP efficacy. Replacing the "loop-beta 8" region (the C-terminal end of the beta-roll) improved agonist sensitivity for cGMP selectively over cAMP. Our results thus identify multiple BD elements outside the C-helix that control selective ligand interaction and channel gating steps by distinct mechanisms. This suggests that the purine ring of the cyclic nucleotide may interact with both the beta-roll and the C-helix at different points in the mechanism.
Collapse
Affiliation(s)
- Edgar C Young
- Center for Neurobiology & Behavior, Columbia University, New York, New York 10032, USA.
| | | |
Collapse
|
35
|
Zagotta WN, Olivier NB, Black KD, Young EC, Olson R, Gouaux E. Structural basis for modulation and agonist specificity of HCN pacemaker channels. Nature 2003; 425:200-5. [PMID: 12968185 DOI: 10.1038/nature01922] [Citation(s) in RCA: 460] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2003] [Accepted: 07/17/2003] [Indexed: 11/08/2022]
Abstract
The family of hyperpolarization-activated, cyclic nucleotide-modulated (HCN) channels are crucial for a range of electrical signalling, including cardiac and neuronal pacemaker activity, setting resting membrane electrical properties and dendritic integration. These nonselective cation channels, underlying the I(f), I(h) and I(q) currents of heart and nerve cells, are activated by membrane hyperpolarization and modulated by the binding of cyclic nucleotides such as cAMP and cGMP. The cAMP-mediated enhancement of channel activity is largely responsible for the increase in heart rate caused by beta-adrenergic agonists. Here we have investigated the mechanism underlying this modulation by studying a carboxy-terminal fragment of HCN2 containing the cyclic nucleotide-binding domain (CNBD) and the C-linker region that connects the CNBD to the pore. X-ray crystallographic structures of this C-terminal fragment bound to cAMP or cGMP, together with equilibrium sedimentation analysis, identify a tetramerization domain and the mechanism for cyclic nucleotide specificity, and suggest a model for ligand-dependent channel modulation. On the basis of amino acid sequence similarity to HCN channels, the cyclic nucleotide-gated, and eag- and KAT1-related families of channels are probably related to HCN channels in structure and mechanism.
Collapse
Affiliation(s)
- William N Zagotta
- Department of Physiology and Biophysics, Howard Hughes Medical Institute, Box 357290, University of Washington School of Medicine, Seattle, Washington 98195-7290, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Peng C, Rich ED, Thor CA, Varnum MD. Functionally important calmodulin-binding sites in both NH2- and COOH-terminal regions of the cone photoreceptor cyclic nucleotide-gated channel CNGB3 subunit. J Biol Chem 2003; 278:24617-23. [PMID: 12730238 DOI: 10.1074/jbc.m301699200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Whereas an important aspect of sensory adaptation in rod photoreceptors and olfactory receptor neurons is thought to be the regulation of cyclic nucleotide-gated (CNG) channel activity by calcium-calmodulin (Ca2+-CaM), it is not clear that cone photoreceptor CNG channels are similarly modulated. Cone CNG channels are composed of at least two different subunit types, CNGA3 and CNGB3. We have investigated whether calmodulin modulates the activity of these channels by direct binding to the CNGB3 subunit. Heteromeric channels were formed by co-expression of human CNGB3 with human CNGA3 subunits in Xenopus oocytes; CNGB3 subunits conferred sensitivity to regulation by Ca2+-CaM, whereas CaM regulation of homomeric CNGA3 channels was not detected. To explore the mechanism underlying this regulation, we localized potential CaM-binding sites in both NH2- and COOH-terminal cytoplasmic domains of CNGB3 using gel-overlay and glutathione S-transferase pull-down assays. For both sites, binding of CaM depended on the presence of Ca2+. Individual deletions of either CaM-binding site in CNGB3 generated channels that remained sensitive to regulation by Ca2+-CaM, but deletion of both together resulted in heteromeric channels that were not modulated. Thus, both NH2- and COOH-terminal CaM-binding sites in CNGB3 are functionally important for regulation of recombinant cone CNG channels. These studies suggest a potential role for direct binding and unbinding of Ca2+-CaM to human CNGB3 during cone photoreceptor adaptation and recovery.
Collapse
Affiliation(s)
- Changhong Peng
- Department of Veterinary and Comparative Anatomy, Washington State University, Pullman 99164-6520, USA
| | | | | | | |
Collapse
|
37
|
Flynn GE, Zagotta WN. A cysteine scan of the inner vestibule of cyclic nucleotide-gated channels reveals architecture and rearrangement of the pore. J Gen Physiol 2003; 121:563-82. [PMID: 12771192 PMCID: PMC2217351 DOI: 10.1085/jgp.200308819] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclic nucleotide-gated (CNG) channels belong to the P-loop-containing family of ion channels that also includes KcsA, MthK, and Shaker channels. In this study, we investigated the structure and rearrangement of the CNGA1 channel pore using cysteine mutations and cysteine-specific modification. We constructed 16 mutant channels, each one containing a cysteine mutation at one of the positions between 384 and 399 in the S6 region of the pore. By measuring currents activated by saturating concentrations of the full agonist cGMP and the partial agonists cIMP and cAMP, we show that mutating S6 residues to cysteine caused both favorable and unfavorable changes in the free energy of channel opening. The time course of cysteine modification with 2-aminoethylmethane thiosulfonate hydrochloride (MTSEA) was complex. For many positions we observed decreases in current activated by cGMP and concomitant increases in current activated by cIMP and cAMP. A model where modification affected both gating and permeation successfully reproduced the complex time course of modification for most of the mutant channels. From the model fits to the time course of modification for each mutant channel, we quantified the following: (a) the bimolecular rate constant of modification in the open state, (b) the change in conductance, and (c) the change in the free energy of channel opening for modification of each cysteine. At many S6 cysteines, modification by MTSEA caused a decrease in conductance and a favorable change in the free energy of channel opening. Our results are interpreted within the structural framework of the known structures of KcsA and MthK. We conclude that: (a) MTSEA modification affects both gating and permeation, (b) the open configuration of the pore of CNGA1 channels is consistent with the structure of MthK, and (c) the modification of S6 residues disrupts the helical packing of the closed channel, making it easier for channels to open.
Collapse
Affiliation(s)
- Galen E Flynn
- Department of Physiology and Biophysics, University of Washington, Seattle 98195-7290, USA
| | | |
Collapse
|
38
|
Trudeau MC, Zagotta WN. Calcium/calmodulin modulation of olfactory and rod cyclic nucleotide-gated ion channels. J Biol Chem 2003; 278:18705-8. [PMID: 12626507 DOI: 10.1074/jbc.r300001200] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic nucleotide-gated (CNG) ion channels mediate sensory transduction in olfactory sensory neurons and retinal photoreceptor cells. In these systems, internal calcium/calmodulin (Ca2+/CaM) inhibits CNG channels, thereby having a putative role in sensory adaptation. Functional differences in Ca2+/CaM-dependent inhibition depend on the different subunit composition of olfactory and rod CNG channels. Recent evidence shows that three subunit types (CNGA2, CNGA4, and CNGB1b) make up native olfactory CNG channels and account for the fast inhibition of native channels by Ca2+/CaM. In contrast, two subunit types (CNGA1 and CNGB1) appear sufficient to mirror the native properties of rod CNG channels, including the inhibition by Ca2+/CaM. Within CNG channel tetramers, specific subunit interactions also mediate Ca2+/CaM-dependent inhibition. In olfactory CNGA2 channels, Ca2+/CaM binds to an N-terminal region and disrupts an interaction between the N- and C-terminal regions, causing inhibition. Ca2+/CaM also binds the N-terminal region of CNGB1 subunits and disrupts an intersubunit, N- and C-terminal interaction between CNGB1 and CNGA1 subunits in rod channels. However, the precise N- and C-terminal regions that form these interactions in olfactory channels are different from those in rod channels. Here, we will review recent advances in understanding the subunit composition and the mechanisms and roles for Ca2+/CaM-dependent inhibition in olfactory and rod CNG channels.
Collapse
Affiliation(s)
- Matthew C Trudeau
- Department of Physiology and Biophysics, Howard Hughes Medical Institute, University of Washington Medical School, Seattle, Washington 98195, USA
| | | |
Collapse
|
39
|
Abstract
By analogy to other cation channel subunits with six transmembrane-spanning domains, the seven members of the "classical" or "canonical" transient receptor potential channels (TRPC) family are believed to assemble into homo- or heterotetrameric complexes. These complexes have been verified by classical methods such as coimmunoprecipitation, crosslinking analysis or functional assays applying dominant negative pore mutants. More recently, fluorescence resonance energy transfer (FRET)-a measure for the close proximity of fluorescent molecules-has become instrumental in monitoring protein assembly in living cells. Here we demonstrate further possibilities and verification procedures of the FRET technology to test the assembly of ion channel subunits. Temporally and spatially resolved FRET imaging demonstrates an early assembly of TRPC subunits in the endoplasmic reticulum and the Golgi apparatus. Confocal FRET imaging verifies FRET signals over the plasma membrane at high spatial resolution. Taking advantage of the quantitative analysis of digital video imaging, we demonstrate that FRET between TRPC subunits is only poorly concentration-dependent. Moreover, a correlation between the efficiency of energy transfer and the molar ratio of the FRET donor to the acceptor was exploited to verify the tetrameric stoichiometry of TRPC complexes. Finally, we introduce a competition-FRET assay to test the ability of wild-type TRPC subunits to recruit fluorescent TRPC subunits into separate channel complexes.
Collapse
Affiliation(s)
- Hemasse Amiri
- Institut für Pharmakologie, Freie Universität Berlin, Thielallee 67-73, 14195 Berlin, Germany
| | | | | |
Collapse
|
40
|
Hoenderop J, Voets T, Hoefs S, Weidema F, Prenen J, Nilius B, Bindels R. Homo- and heterotetrameric architecture of the epithelial Ca2+ channels TRPV5 and TRPV6. EMBO J 2003; 22:776-85. [PMID: 12574114 PMCID: PMC145440 DOI: 10.1093/emboj/cdg080] [Citation(s) in RCA: 246] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The molecular assembly of the epithelial Ca(2+) channels (TRPV5 and TRPV6) was investigated to determine the subunit stoichiometry and composition. Immunoblot analysis of Xenopus laevis oocytes expressing TRPV5 and TRPV6 revealed two specific bands of 75 and 85-100 kDa, corresponding to the core and glycosylated proteins, respectively, for each channel. Subsequently, membranes of these oocytes were sedimented on sucrose gradients. Immuno blotting revealed that TRPV5 and TRPV6 complexes migrate with a mol. wt of 400 kDa, in line with a tetrameric structure. The tetrameric stoichiometry was confirmed in an electrophysiological analysis of HEK293 cells co-expressing concatemeric channels together with a TRPV5 pore mutant that reduced Cd(2+) sensitivity and voltage-dependent gating. Immuno precipitations using membrane fractions from oocytes co-expressing TRPV5 and TRPV6 demonstrated that both channels can form heteromeric complexes. Expression of all possible heterotetrameric TRPV5/6 complexes in HEK293 cells resulted in Ca(2+) channels that varied with respect to Ca(2+)-dependent inactivation, Ba(2+) selectivity and pharmacological block. Thus, Ca(2+)-transporting epithelia co-expressing TRPV5 and TRPV6 can generate a pleiotropic set of functional heterotetrameric channels with different Ca(2+) transport kinetics.
Collapse
Affiliation(s)
| | - T. Voets
- Department of Cell Physiology, Nijmegen Centre for Molecular Life Sciences, University Medical Centre Nijmegen, PO Box 9101, NL-6500 HB Nijmegen, The Netherlands and
Department of Physiology, Campus Gasthuisberg, KU Leuven, Belgium Corresponding author e-mail:
| | | | | | - J. Prenen
- Department of Cell Physiology, Nijmegen Centre for Molecular Life Sciences, University Medical Centre Nijmegen, PO Box 9101, NL-6500 HB Nijmegen, The Netherlands and
Department of Physiology, Campus Gasthuisberg, KU Leuven, Belgium Corresponding author e-mail:
| | - B. Nilius
- Department of Cell Physiology, Nijmegen Centre for Molecular Life Sciences, University Medical Centre Nijmegen, PO Box 9101, NL-6500 HB Nijmegen, The Netherlands and
Department of Physiology, Campus Gasthuisberg, KU Leuven, Belgium Corresponding author e-mail:
| | - R.J.M. Bindels
- Department of Cell Physiology, Nijmegen Centre for Molecular Life Sciences, University Medical Centre Nijmegen, PO Box 9101, NL-6500 HB Nijmegen, The Netherlands and
Department of Physiology, Campus Gasthuisberg, KU Leuven, Belgium Corresponding author e-mail:
| |
Collapse
|
41
|
Abstract
Cyclic nucleotide-gated channels are key components in the transduction of visual and olfactory signals where their role is to respond to changes in the intracellular concentration of cyclic nucleotides. Although these channels poorly select between physiologically relevant monovalent cations, the gating by cyclic nucleotide is different in the presence of Na(+) or K(+) ions. This property was investigated using rod cyclic nucleotide-gated channels formed by expressing the subunit 1 (or alpha) in HEK293 cells. In the presence of K(+) as the permeant ion, the affinity for cGMP is higher than the affinity measured in the presence of Na(+). At the single channel level, subsaturating concentrations of cGMP show that the main effect of the permeant K(+) ions is to prolong the time channels remain open without major changes in the shut time distribution. In addition, the maximal open probability was higher when K(+) was the permeant ion (0.99 for K(+) vs. 0.95 for Na(+)) due to an increase in the apparent mean open time. Similarly, in the presence of saturating concentrations of cAMP, known to bind but unable to efficiently open the channel, permeant K(+) ions also prolong the time channels visit the open state. Together, these results suggest that permeant ions alter the stability of the open conformation by influencing of the O-->C transition.
Collapse
Affiliation(s)
- Miguel Holmgren
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
42
|
Nicke A, Rettinger J, Schmalzing G. Monomeric and dimeric byproducts are the principal functional elements of higher order P2X1 concatamers. Mol Pharmacol 2003; 63:243-52. [PMID: 12488557 DOI: 10.1124/mol.63.1.243] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Heteromultimeric assembly of ion channel subunits generates high diversity in ion channel subtypes with distinct pharmacological and functional properties. To determine the subunit stoichiometry and order of ion channels, constructs with several concatenated subunits have been widely used in electrophysiological studies. Here we used primarily biochemical techniques to analyze the synthesis, assembly, and surface expression of P2X1 concatamers. We found that full-length concatamers consisting of two to six contiguous copies of the P2X1 subunit, although readily synthesized in Xenopus laevis oocytes, were entirely retained as aggregates in the endoplasmic reticulum. In contrast, minute levels of lower order byproducts, such as monomers and dimers, that were inherently formed with all the concatamers combined to form defined protein complexes equal in mass to the homotrimeric P2X1 receptor assembled from P2X1 monomers. Besides these complexes consisting of three monomers or one monomer plus one concatenated dimer, only small amounts of concatenated P2X1 trimers reached the plasma membrane. Complexes comprising more than three subunits were not observed in the plasma membrane. The byproduct complexes can account fully for the ATP-gated currents arising from expression of concatenated P2X1 subunits. These results strongly corroborate a trimeric architecture for P2X receptors yet indicate that formation of lower order by-products can be a pitfall of the concatamer approach.
Collapse
Affiliation(s)
- Annette Nicke
- Department of Molecular Pharmacology, Medical School of the Technical University of Aachen, Aachen, Germany
| | | | | |
Collapse
|
43
|
Abstract
Members of the voltage-gated family of ion channels generally demonstrate rotational symmetry about their pore regions. Recent evidence suggests that a subset of this family, the cyclic nucleotide-gated channels, may deviate from this pattern of rotational symmetry by having 3A:1B subunit stoichiometry. This finding raises many questions about the function, assembly, and trafficking of these and related ion channels and about the functional nonequivalence of subunits with identical amino acid sequences.
Collapse
Affiliation(s)
- Anita L Zimmerman
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
44
|
Weitz D, Ficek N, Kremmer E, Bauer PJ, Kaupp UB. Subunit stoichiometry of the CNG channel of rod photoreceptors. Neuron 2002; 36:881-9. [PMID: 12467591 DOI: 10.1016/s0896-6273(02)01098-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cyclic nucleotide-gated (CNG) channels play a central role in the conversion of sensory stimuli into electrical signals. CNG channels form heterooligomeric complexes built of A and B subunits. Here, we study the subunit stoichiometry of the native rod CNG channel by chemical crosslinking. The apparent molecular weight (M(w)) of each crosslink product was determined by SDS-PAGE, and its composition was analyzed by Western blotting using antibodies specific for the A1 or B1 subunit. The number of crosslink products and their M(w) as well as the immunological identification of A1 and B1 subunits in the crosslink products led us to conclude that the native rod CNG channel is a tetramer composed of three A1 and one B1 subunit. This is an example of violation of symmetry in tetrameric channels.
Collapse
Affiliation(s)
- Dietmar Weitz
- Institut für Biologische Informationsverarbeitung, Forschungszentrum Jülich, Germany
| | | | | | | | | |
Collapse
|
45
|
Mazzolini M, Punta M, Torre V. Movement of the C-helix during the gating of cyclic nucleotide-gated channels. Biophys J 2002; 83:3283-95. [PMID: 12496096 PMCID: PMC1302404 DOI: 10.1016/s0006-3495(02)75329-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Movements within the cyclic nucleotide-binding domain of cyclic nucleotide-gated channels are thought to underlie the initial phase of channel gating (Tibbs, G. R., D. T. Liu, B. G. Leypold, and S. A. Siegelbaum. 1998. J. Biol. Chem. 273:4497-4505; Zong, X., H. Zucker, F. Hofmann, and M. Biel. 1998. EMBO J. 17:353-362; Matulef, K., G. E. Flynn, and W. N. Zagotta. 1999. Neuron. 24:443-452; Paoletti, P., E. C. Young, and S. A. Siegelbaum. 1999. J. Gen. Physiol. 113:17-33; Johnson, J. P., and W. N. Zagotta. 2001. Nature. 412:917-921). To investigate these movements, cysteine mutation was performed on each of the 28 residues (Leu-583 to Asn-610), which span the agonist-binding domain of the alpha-subunit of the bovine rod cyclic nucleotide-gated channel. The effects of Cd(2+) ions, 2-trimethylammonioethylmethane thiosulfonate (MTSET) and copper phenanthroline (CuP) on channel activity were examined, in excised inside-out patches in the presence and in the absence of a saturating concentration of cGMP. The application of 100 microM Cd(2+) in the presence of saturating concentration of cGMP caused an irreversible and almost complete reduction of the current in mutant channels E594C, I600C, and L601C. In the absence of cGMP, the presence of 100 microM Cd(2+) caused a strong current reduction in all cysteine mutants from Asp-588 to Leu-607, with the exception of mutant channels A589C, M592C, M602C, K603C, and L606C. The selective effect of Cd(2+) ions was very similar to that observed when adding the oxidizing agent CuP to the bath medium, except for mutant channel G597C, where CuP caused a stronger current decrease (67 +/- 7%) than Cd(2+) (23 +/- 4%). In the absence of cGMP, MTSET caused a reduction of the current by >40% in mutant channels L607C, L601C, I600C, G597C, and E594C, whereas in the presence of cGMP only mutant channel L601C was affected. The application of MTSET protected many mutant channels from the effects of Cd(2+) and CuP. These results suggest that, when CNG channels are in the open state, residues from Asp-588 to Leu-607 are in an alpha-helical structure, homologous to the C-helix of the catabolite gene activator protein (Weber, I. T., and T. A. Steitz. 1987. J. Mol. Biol. 198:311-326). Furthermore, residues Glu-594, Gly-597, Ile-600, and Leu-601 of these helices belonging to two different subunits must be in close proximity. In the closed state the C-helices are in a different configuration and undergo significant fluctuations.
Collapse
Affiliation(s)
- Monica Mazzolini
- INFM Section and International School for Advanced Studies, via Beirut 2-4, I-34014 Trieste, Italy
| | | | | |
Collapse
|
46
|
Kwon RJ, Ha TS, Kim W, Park CS. Binding symmetry of extracellular divalent cations to conduction pore studied using tandem dimers of a CNG channel. Biochem Biophys Res Commun 2002; 298:478-85. [PMID: 12408977 DOI: 10.1016/s0006-291x(02)02507-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cyclic nucleotide-gated (CNG) channels are composed of the tetramer of alpha-subunit alone or alpha- and beta-subunits. The alpha-subunits of these channels have a conserved glutamate (Glu) residue within the pore-forming region and the residue determines the selectivity as well as the affinity for the extracellular divalent cations. Using the high-affinity mutant (E363D) of bovine retinal CNG channel in which the Glu at position 363 was replaced to Asp, we constructed tandem dimers and investigated the binding characteristics of divalent cations to the site. The gating and permeation characteristics of individual homomeric tandem dimers are indistinguishable to those of homo-tetramers formed by parental monomers. The heteromeric tandem dimers showed the binding affinity for Sr(2+) identical to the geometric mean of the affinities for two parent channels, indicating the energy additive and thus the simultaneous interaction. On the other hand, the binding affinity for Mg(2+) followed the harmonic mean of those parent channels indicating that Mg(2+) interacts more strongly with the subunit bearing Asp residue at the position. Thus the results strongly suggest that the Glu363 residues in the CNG channel pore be flexible enough to adapt different binding symmetries for different divalent cations. Moreover, the simultaneous interaction between the four Glu residues and Sr(2+) provides an important structural constraint to the CNG channel outer vestibule of unknown structure.
Collapse
Affiliation(s)
- Ryuk-Jun Kwon
- Laboratory of Molecular Neurobiology, Department of Life Science, Kwangju Institute of Science and Technology (K-JIST), 1 Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea
| | | | | | | |
Collapse
|
47
|
Decressac S, Grechez-Cassiau A, Lenfant J, Falcón J, Bois P. Cloning, localization and functional properties of a cGMP-gated channel in photoreceptor cells from fish pineal gland. J Pineal Res 2002; 33:225-33. [PMID: 12390505 DOI: 10.1034/j.1600-079x.2002.02922.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The perception of photic information and its translation into a rhythmic melatonin signal differ considerably among vertebrates. In the fish pineal gland, melatonin biosynthesis is controlled directly by the natural light/dark cycle. There are indications that the mechanisms of phototransduction are similar in the retinal and pineal photoreceptor cells. Here we report the molecular cloning of a novel ionic cyclic guanosine monophosphate (cGMP)-gated channel from trout pineal photoreceptors. The deduced amino acid sequence exhibits a high sequence homology to cyclic nucleotide-gated-3 (CNG) channels from retinal cones. In situ hybridization with sections of trout pineal gland revealed the expression of CNG channel in photoreceptor cells of the pineal organ. Electrophysiological studies by means of patch-clamp technique indicated that the native channel in photoreceptor cells and the expressed channel in a human cell line (HEK 293 cells) have properties similar to those of cone-CNG (cCNG)-3 channels. They are activated by cGMP, insensitive to cyclic adenosine monophosphate (cAMP) and blocked by intracellular Mg2+ ions at positive voltage values. They have a single-channel conductance close to 42 pS in negative voltage range. In transfected HEK cells loaded with the calcium indicator dye Fura 2, direct activation of CNG channels by 8-Br-cGMP increased fluorescence. The signal was blocked by the addition of Mg2+ ions. From these results, it is suggested that the pineal cyclic nucleotide-gated channel is a good candidate for mediating calcium entry into the pineal photoreceptors. It is most probably a key element in the signalling pathways that control the rhythmic production of melatonin.
Collapse
Affiliation(s)
- Sonia Decressac
- Laboratoire des Biomembranes et Signalisation Cellulaire, Unité Mixte de Recherche CNRS 6558, Université de Poitiers, 40 Avenue du Recteur Pineau, 86022 Poitiers Cedex, France
| | | | | | | | | |
Collapse
|
48
|
Abstract
Cyclic nucleotide-gated (CNG) channels comprise four subunits and are activated by the direct binding of cyclic nucleotide to an intracellular domain on each subunit. This ligand binding domain is thought to contain a beta roll followed by two alpha helices, designated the B and C helices. To examine the quaternary structure of CNG channels and how it changes during ion channel gating, we introduced single cysteines along the C helix of each subunit in an otherwise cysteineless channel. We found that cysteines on the C helices could form intersubunit disulfide bonds, even between diagonal subunits. Disulfide bond formation occurred primarily in closed channels and inhibited channel opening. These data suggest that the C helices from all four channel subunits are in close proximity in the closed state and move apart during channel opening.
Collapse
Affiliation(s)
- Kimberly Matulef
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
49
|
|
50
|
Bennett N, Ildefonse M, Pagès F, Ragno M. Gating of heteromeric retinal rod channels by cyclic AMP: role of the C-terminal and pore domains. Biophys J 2002; 83:920-31. [PMID: 12124274 PMCID: PMC1302196 DOI: 10.1016/s0006-3495(02)75218-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cyclic nucleotide-gated channels are tetramers composed of homologous alpha and beta subunits. C-terminal truncation mutants of the alpha and beta subunits of the retinal rod channel were expressed in Xenopus oocytes, and analyzed for cGMP- and cAMP-induced currents (single-channel records and macroscopic currents). When the alpha subunit truncated downstream of the cGMP-binding site (alpha D608stop) is co-injected with truncated beta subunits, the heteromeric channels present a drastic increase of cAMP sensitivity. A partial effect is observed with heteromeric alpha R656stop-containing channels, while alpha K665stop-containing channels behave like alpha wt/beta wt. The three truncated alpha subunits have wild-type activity when expressed alone. Heteromeric channels composed of alpha wt or truncated alpha subunits and chimeric beta subunits containing the pore domain of the alpha subunit have the same cAMP sensitivity as alpha-only channels. The results disclose the key role of two domains distinct from the nucleotide binding site in the gating of heteromeric channels by cAMP: the pore of the beta subunit, which has an activating effect, and a conserved domain situated downstream of the cGMP-binding site in the alpha subunit (I609-K665), which inhibits this effect.
Collapse
Affiliation(s)
- Nelly Bennett
- Département de Biologie Moléculaire et Structurale, Laboratoire BMC, UMR CNRS 5090, CEA-Grenoble, Grenoble, France.
| | | | | | | |
Collapse
|