Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
bioRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-37732182

ABSTRACT

Cell membranes consist of heterogeneous lipid domains that influence key cellular processes, including signal transduction, endocytosis, and electrical excitability. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we found that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. The size of OMDs is also dependent on cholesterol levels and the structure of lipid tails. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. This increased neuronal firing could be partially due to an increased open probability of HCN channels. In animal models of neuropathic pain, we observed shrunken OMDs and relocalization of HCN channels from OMDs to disordered lipid domains. The gating effect on HCN channels was likely a result of direct modulation of the voltage sensor by OMDs. These findings suggest that disturbances in lipid domains play a role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.

2.
Nat Commun ; 14(1): 6595, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37852983

ABSTRACT

Ion channels function within a membrane environment characterized by dynamic lipid compartmentalization. Limited knowledge exists regarding the response of voltage-gated ion channels to transmembrane potential within distinct membrane compartments. By leveraging fluorescence lifetime imaging microscopy (FLIM) and Förster resonance energy transfer (FRET), we visualized the localization of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in membrane domains. HCN4 exhibits a greater propensity for incorporation into ordered lipid domains compared to HCN1. To investigate the conformational changes of the S4 helix voltage sensor of HCN channels, we used dual stop-codon suppression to incorporate different noncanonical amino acids, orthogonal click chemistry for site-specific fluorescence labeling, and transition metal FLIM-FRET. Remarkably, altered FRET levels were observed between VSD sites within HCN channels upon disruption of membrane domains. We propose that the voltage-sensor rearrangements, directly influenced by membrane lipid domains, can explain the heightened activity of pacemaker HCN channels when localized in cholesterol-poor, disordered lipid domains, leading to membrane hyperexcitability and diseases.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membrane Lipids , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Cyclic Nucleotide-Gated Cation Channels/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism
3.
Mo Med ; 120(5): 367-373, 2023.
Article in English | MEDLINE | ID: mdl-37841571

ABSTRACT

Charged ions and ion channels play a critical role in regulating the electrical activities of excitable cells. This review discusses the principles of ion channel regulation in the time domain, as well as the diseases that can arise from channel dysfunction and disturbances in ionic balance. Ion channel signaling is a dynamic process that is essential for various physiological functions, including pain sensation, motor control, and the body's response to stress, such as fight-or-flight response.


Subject(s)
Channelopathies , Humans , Channelopathies/metabolism , Ion Channels/metabolism
4.
J Gen Physiol ; 154(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35583815

ABSTRACT

Phosphoinositide membrane lipids are ubiquitous low-abundance signaling molecules. They direct many physiological processes that involve ion channels, membrane identification, fusion of membrane vesicles, and vesicular endocytosis. Pools of these lipids are continually broken down and refilled in living cells, and the rates of some of these reactions are strongly accelerated by physiological stimuli. Recent biophysical experiments described here measure and model the kinetics and regulation of these lipid signals in intact cells. Rapid on-line monitoring of phosphoinositide metabolism is made possible by optical tools and electrophysiology. The experiments reviewed here reveal that as for other cellular second messengers, the dynamic turnover and lifetimes of membrane phosphoinositides are measured in seconds, controlling and timing rapid physiological responses, and the signaling is under strong metabolic regulation. The underlying mechanisms of this metabolic regulation remain questions for the future.


Subject(s)
Endocytosis , Phosphatidylinositols , Lipid Metabolism , Phosphatidylinositols/metabolism , Protein Transport , Signal Transduction
5.
J Gen Physiol ; 154(4)2022 04 04.
Article in English | MEDLINE | ID: mdl-35201266

ABSTRACT

ß-Secretase 1 (ß-site amyloid precursor protein [APP]-cleaving enzyme 1, BACE1) plays a crucial role in the amyloidogenesis of Alzheimer's disease (AD). BACE1 was also discovered to act like an auxiliary subunit to modulate neuronal KCNQ2/3 channels independently of its proteolytic function. BACE1 is palmitoylated at its carboxyl-terminal region, which brings BACE1 to ordered, cholesterol-rich membrane microdomains (lipid rafts). However, the physiological consequences of this specific localization of BACE1 remain elusive. Using spectral Förster resonance energy transfer (FRET), BACE1 and KCNQ2/3 channels were confirmed to form a signaling complex, a phenomenon that was relatively independent of the palmitoylation of BACE1. Nevertheless, palmitoylation of BACE1 was required for recruitment of KCNQ2/3 channels to lipid-raft domains. Two fluorescent probes, designated L10 and S15, were used to label lipid-raft and non-raft domains of the plasma membrane, respectively. Coexpressing BACE1 substantially elevated FRET between L10 and KCNQ2/3, whereas the BACE1-4C/A quadruple mutation failed to produce this effect. In contrast, BACE1 had no significant effect on FRET between S15 probes and KCNQ2/3 channels. A reduction of BACE1-dependent FRET between raft-targeting L10 probes and KCNQ2/3 channels by applying the cholesterol-extracting reagent methyl-ß-cyclodextrin (MßCD), raft-disrupting general anesthetics, or pharmacological inhibitors of palmitoylation, all supported the hypothesis of the palmitoylation-dependent and raft-specific localization of KCNQ2/3 channels. Furthermore, mutating the four carboxyl-terminal cysteines (4C/A) of BACE1 abolished the BACE1-dependent increase of FRET between KCNQ2/3 and the lipid raft-specific protein caveolin 1. Taking these data collectively, we propose that the AD-related protein BACE1 underlies the localization of a neuronal potassium channel.


Subject(s)
Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Cholesterol , Lipoylation/physiology , Membrane Microdomains/metabolism
7.
Nat Commun ; 12(1): 2802, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990563

ABSTRACT

Pacemaker hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels exhibit a reversed voltage-dependent gating, activating by membrane hyperpolarization instead of depolarization. Sea urchin HCN (spHCN) channels also undergo inactivation with hyperpolarization which occurs only in the absence of cyclic nucleotide. Here we applied transition metal ion FRET, patch-clamp fluorometry and Rosetta modeling to measure differences in the structural rearrangements between activation and inactivation of spHCN channels. We found that removing cAMP produced a largely rigid-body rotation of the C-linker relative to the transmembrane domain, bringing the A' helix of the C-linker in close proximity to the voltage-sensing S4 helix. In addition, rotation of the C-linker was elicited by hyperpolarization in the absence but not the presence of cAMP. These results suggest that - in contrast to electromechanical coupling for channel activation - the A' helix serves to couple the S4-helix movement for channel inactivation, which is likely a conserved mechanism for CNBD-family channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Animals , Cyclic AMP , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/metabolism , Female , Fluorescence Resonance Energy Transfer , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Ion Channel Gating , Male , Mechanotransduction, Cellular , Membrane Potentials , Models, Molecular , Oocytes/metabolism , Patch-Clamp Techniques , Protein Conformation, alpha-Helical , Protein Domains , Sea Urchins/metabolism , Spermatozoa/metabolism , Xenopus/metabolism
8.
Nat Struct Mol Biol ; 26(8): 686-694, 2019 08.
Article in English | MEDLINE | ID: mdl-31285608

ABSTRACT

Voltage-gated ion channels (VGICs) contain positively charged residues within the S4 helix of the voltage-sensing domain (VSD) that are displaced in response to changes in transmembrane voltage, promoting conformational changes that open the pore. Pacemaker hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are unique among VGICs because their open probability is increased by membrane hyperpolarization rather than depolarization. Here we measured the precise movement of the S4 helix of a sea urchin HCN channel using transition metal ion fluorescence resonance energy transfer (tmFRET). We show that the S4 undergoes a substantial (~10 Å) downward movement in response to membrane hyperpolarization. Furthermore, by applying distance constraints determined from tmFRET experiments to Rosetta modeling, we reveal that the carboxy-terminal part of the S4 helix exhibits an unexpected tilting motion during hyperpolarization activation. These data provide a long-sought glimpse of the hyperpolarized state of a functioning VSD and also a framework for understanding the dynamics of reverse gating in HCN channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Amino Acid Sequence , Animals , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Ion Channel Gating/physiology , Membrane Potentials , Models, Molecular , Motion , Patch-Clamp Techniques , Point Mutation , Potassium/metabolism , Protein Conformation , Protein Domains , Recombinant Proteins/chemistry , Strongylocentrotus purpuratus/chemistry
9.
J Gen Physiol ; 150(4): 625-635, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29567795

ABSTRACT

KCNH voltage-gated potassium channels (EAG, ERG, and ELK) play significant roles in neuronal and cardiac excitability. They contain cyclic nucleotide-binding homology domains (CNBHDs) but are not directly regulated by cyclic nucleotides. Instead, the CNBHD ligand-binding cavity is occupied by an intrinsic ligand, which resides at the intersubunit interface between the N-terminal eag domain and the C-terminal CNBHD. We show that, in Danio rerio ELK channels, this intrinsic ligand is critical for voltage-dependent potentiation (VDP), a process in which channel opening is stabilized by prior depolarization. We demonstrate that an exogenous peptide corresponding to the intrinsic ligand can bind to and regulate zebrafish ELK channels. This exogenous intrinsic ligand inhibits the channels before VDP and potentiates the channels after VDP. Furthermore, using transition metal ion fluorescence resonance energy transfer and a fluorescent noncanonical amino acid L-Anap, we show that there is a rearrangement of the intrinsic ligand relative to the CNBHD during VDP. We propose that the intrinsic ligand switches from antagonist to agonist as a result of a rearrangement of the eag domain-CNBHD interaction during VDP.


Subject(s)
Membrane Transport Modulators/pharmacology , Peptides/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Zebrafish Proteins/metabolism , Action Potentials , Animals , Binding Sites , Ligands , Peptides/chemistry , Potassium Channels, Voltage-Gated/chemistry , Protein Binding , Xenopus , Zebrafish , Zebrafish Proteins/chemistry
10.
J Gen Physiol ; 150(2): 211-224, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29326133

ABSTRACT

Lipids influence powerfully the function of ion channels and transporters in two well-documented ways. A few lipids act as bona fide second messengers by binding to specific sites that control channel and transporter gating. Other lipids act nonspecifically by modifying the physical environment of channels and transporters, in particular the protein-membrane interface. In this short review, we first consider lipid signaling from this traditional viewpoint, highlighting innumerable Journal of General Physiology publications that have contributed to our present understanding. We then switch to our own emerging view that much important lipid signaling occurs via the formation of membrane domains that influence the function of channels and transporters within them, promote selected protein-protein interactions, and control the turnover of surface membrane.


Subject(s)
Endocytosis , Membrane Lipids/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Second Messenger Systems , Animals , Humans
12.
Elife ; 62017 04 27.
Article in English | MEDLINE | ID: mdl-28443815

ABSTRACT

EAG-like (ELK) voltage-gated potassium channels are abundantly expressed in the brain. These channels exhibit a behavior called voltage-dependent potentiation (VDP), which appears to be a specialization to dampen the hyperexitability of neurons. VDP manifests as a potentiation of current amplitude, hyperpolarizing shift in voltage sensitivity, and slowing of deactivation in response to a depolarizing prepulse. Here we show that VDP of D. rerio ELK channels involves the structural interaction between the intracellular N-terminal eag domain and C-terminal CNBHD. Combining transition metal ion FRET, patch-clamp fluorometry, and incorporation of a fluorescent noncanonical amino acid, we show that there is a rearrangement in the eag domain-CNBHD interaction with the kinetics, voltage-dependence, and ATP-dependence of VDP. We propose that the activation of ELK channels involves a slow open-state dependent rearrangement of the direct interaction between the eag domain and CNBHD, which stabilizes the opening of the channel.


Subject(s)
Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Fluorescence Resonance Energy Transfer , Fluorometry , Kinetics , Models, Molecular , Patch-Clamp Techniques , Protein Conformation , Protein Domains , Zebrafish
13.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 513-522, 2017 May.
Article in English | MEDLINE | ID: mdl-28189644

ABSTRACT

Phosphoinositides are rapidly turning-over phospholipids that play key roles in intracellular signaling and modulation of membrane effectors. Through technical refinements we have improved sensitivity in the analysis of the phosphoinositide PI, PIP, and PIP2 pools from living cells using mass spectrometry. This has permitted further resolution in phosphoinositide lipidomics from cell cultures and small samples of tissue. The technique includes butanol extraction, derivatization of the lipids, post-column infusion of sodium to stabilize formation of sodiated adducts, and electrospray ionization mass spectrometry in multiple reaction monitoring mode, achieving a detection limit of 20pg. We describe the spectrum of fatty-acyl chains in the cellular phosphoinositides. Consistent with previous work in other mammalian primary cells, the 38:4 fatty-acyl chains dominate in the phosphoinositides of the pineal gland and of superior cervical ganglia, and many additional fatty acid combinations are found at low abundance. However, Chinese hamster ovary cells and human embryonic kidney cells (tsA201) in culture have different fatty-acyl chain profiles that change with growth state. Their 38:4 lipids lose their dominance as cultures approach confluence. The method has good time resolution and follows well the depletion in <20s of both PIP2 and PIP that results from strong activation of Gq-coupled receptors. The receptor-activated phospholipase C exhibits no substrate selectivity among the various fatty-acyl chain combinations.


Subject(s)
Fatty Acids/isolation & purification , Phosphatidylinositols/isolation & purification , Phospholipids/isolation & purification , Spectrometry, Mass, Electrospray Ionization/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Fatty Acids/chemistry , Humans , Phosphatidylinositols/chemistry , Phospholipids/chemistry , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 113(23): E3290-9, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27217553

ABSTRACT

Myo-inositol is an important cellular osmolyte in autoregulation of cell volume and fluid balance, particularly for mammalian brain and kidney cells. We find it also regulates excitability. Myo-inositol is the precursor of phosphoinositides, key signaling lipids including phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]. However, whether myo-inositol accumulation during osmoregulation affects signaling and excitability has not been fully explored. We found that overexpression of the Na(+)/myo-inositol cotransporter (SMIT1) and myo-inositol supplementation enlarged intracellular PI(4,5)P2 pools, modulated several PI(4,5)P2-dependent ion channels including KCNQ2/3 channels, and attenuated the action potential firing of superior cervical ganglion neurons. Further experiments using the rapamycin-recruitable phosphatase Sac1 to hydrolyze PI(4)P and the P4M probe to visualize PI(4)P suggested that PI(4)P levels increased after myo-inositol supplementation with SMIT1 expression. Elevated relative levels of PIP and PIP2 were directly confirmed using mass spectrometry. Inositol trisphosphate production and release of calcium from intracellular stores also were augmented after myo-inositol supplementation. Finally, we found that treatment with a hypertonic solution mimicked the effect we observed with SMIT1 overexpression, whereas silencing tonicity-responsive enhancer binding protein prevented these effects. These results show that ion channel function and cellular excitability are under regulation by several "physiological" manipulations that alter the PI(4,5)P2 setpoint. We demonstrate a previously unrecognized linkage between extracellular osmotic changes and the electrical properties of excitable cells.


Subject(s)
Heat-Shock Proteins/metabolism , Neurons/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Symporters/metabolism , Action Potentials , Calcium Signaling , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , HEK293 Cells , Heat-Shock Proteins/genetics , Humans , Inositol/metabolism , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/metabolism , Osmoregulation , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Symporters/genetics , TRPM Cation Channels/metabolism , Transcription Factors/metabolism
15.
J Biol Chem ; 289(19): 13680-90, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24675082

ABSTRACT

Precursor mRNA encoding CNGA3 subunits of cone photoreceptor cyclic nucleotide-gated (CNG) channels undergoes alternative splicing, generating isoforms differing in the N-terminal cytoplasmic region of the protein. In humans, four variants arise from alternative splicing, but the functional significance of these changes has been a persistent mystery. Heterologous expression of the four possible CNGA3 isoforms alone or with CNGB3 subunits did not reveal significant differences in basic channel properties. However, inclusion of optional exon 3, with or without optional exon 5, produced heteromeric CNGA3 + CNGB3 channels exhibiting an ∼2-fold greater shift in K1/2,cGMP after phosphatidylinositol 4,5-biphosphate or phosphatidylinositol 3,4,5-trisphosphate application compared with channels lacking the sequence encoded by exon 3. We have previously identified two structural features within CNGA3 that support phosphoinositides (PIPn) regulation of cone CNG channels: N- and C-terminal regulatory modules. Specific mutations within these regions eliminated PIPn sensitivity of CNGA3 + CNGB3 channels. The exon 3 variant enhanced the component of PIPn regulation that depends on the C-terminal region rather than the nearby N-terminal region, consistent with an allosteric effect on PIPn sensitivity because of altered N-C coupling. Alternative splicing of CNGA3 occurs in multiple species, although the exact variants are not conserved across CNGA3 orthologs. Optional exon 3 appears to be unique to humans, even compared with other primates. In parallel, we found that a specific splice variant of canine CNGA3 removes a region of the protein that is necessary for high sensitivity to PIPn. CNGA3 alternative splicing may have evolved, in part, to tune the interactions between cone CNG channels and membrane-bound phosphoinositides.


Subject(s)
Alternative Splicing/physiology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Exons , Eye Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Animals , Cyclic Nucleotide-Gated Cation Channels/genetics , Dogs , Evolution, Molecular , Eye Proteins/genetics , Female , Humans , Male , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol Phosphates/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retinal Cone Photoreceptor Cells/cytology
16.
Am J Physiol Cell Physiol ; 305(2): C147-59, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23552282

ABSTRACT

Cyclic nucleotide-gated (CNG) channels are critical for sensory transduction in retinal photoreceptors and olfactory receptor cells; their activity is modulated by phosphoinositides (PIPn) such as phosphatidylinositol 4,5-bisphosphate (PIP2) and phosphatidylinositol 3,4,5-trisphosphate (PIP3). An achromatopsia-associated mutation in cone photoreceptor CNGA3, L633P, is located in a carboxyl (COOH)-terminal leucine zipper domain shown previously to be important for channel assembly and PIPn regulation. We determined the functional consequences of this mutation using electrophysiological recordings of patches excised from cells expressing wild-type and mutant CNG channel subunits. CNGA3-L633P subunits formed functional channels with or without CNGB3, producing an increase in apparent cGMP affinity. Surprisingly, L633P dramatically potentiated PIPn inhibition of apparent cGMP affinity for these channels. The impact of L633P on PIPn sensitivity depended on an intact amino (NH2) terminal PIPn regulation module. These observations led us to hypothesize that L633P enhances PIPn inhibition by altering the coupling between NH2- and COOH-terminal regions of CNGA3. A recombinant COOH-terminal fragment partially restored normal PIPn sensitivity to channels with COOH-terminal truncation, but L633P prevented this effect. Furthermore, coimmunoprecipitation of channel fragments, and thermodynamic linkage analysis, also provided evidence for NH2-COOH interactions. Finally, tandem dimers of CNGA3 subunits that specify the arrangement of subunits containing L633P and other mutations indicated that the putative interdomain interaction occurs between channel subunits (intersubunit) rather than exclusively within the same subunit (intrasubunit). Collectively, these studies support a model in which intersubunit interactions control the sensitivity of cone CNG channels to regulation by phosphoinositides. Aberrant channel regulation may contribute to disease progression in patients with the L633P mutation.


Subject(s)
Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/metabolism , Gene Expression Regulation/physiology , Phosphatidylinositols/pharmacology , Protein Subunits , Retinal Cone Photoreceptor Cells/drug effects , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/genetics , Humans , Mutagenesis, Site-Directed , Oocytes/metabolism , Point Mutation , Retinal Cone Photoreceptor Cells/metabolism , Xenopus laevis
17.
J Gen Physiol ; 141(4): 413-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23530136

ABSTRACT

Cyclic nucleotide-gated (CNG) channels in retinal photoreceptors play a crucial role in vertebrate phototransduction. The ligand sensitivity of photoreceptor CNG channels is adjusted during adaptation and in response to paracrine signals, but the mechanisms involved in channel regulation are only partly understood. Heteromeric cone CNGA3 (A3) + CNGB3 (B3) channels are inhibited by membrane phosphoinositides (PIP(n)), including phosphatidylinositol 3,4,5-triphosphate (PIP(3)) and phosphatidylinositol 4,5-bisphosphate (PIP(2)), demonstrating a decrease in apparent affinity for cyclic guanosine monophosphate (cGMP). Unlike homomeric A1 or A2 channels, A3-only channels paradoxically did not show a decrease in apparent affinity for cGMP after PIP(n) application. However, PIP(n) induced an ∼2.5-fold increase in cAMP efficacy for A3 channels. The PIP(n)-dependent change in cAMP efficacy was abolished by mutations in the C-terminal region (R643Q/R646Q) or by truncation distal to the cyclic nucleotide-binding domain (613X). In addition, A3-613X unmasked a threefold decrease in apparent cGMP affinity with PIP(n) application to homomeric channels, and this effect was dependent on conserved arginines within the N-terminal region of A3. Together, these results indicate that regulation of A3 subunits by phosphoinositides exhibits two separable components, which depend on structural elements within the N- and C-terminal regions, respectively. Furthermore, both N and C regulatory modules in A3 supported PIP(n) regulation of heteromeric A3+B3 channels. B3 subunits were not sufficient to confer PIP(n) sensitivity to heteromeric channels formed with PIP(n)-insensitive A subunits. Finally, channels formed by mixtures of PIP(n)-insensitive A3 subunits, having complementary mutations in N- and/or C-terminal regions, restored PIP(n) regulation, implying that intersubunit N-C interactions help control the phosphoinositide sensitivity of cone CNG channels.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Binding Sites , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/genetics , Humans , Ion Channel Gating , Molecular Sequence Data , Mutation, Missense , Protein Multimerization , Protein Structure, Tertiary , Protein Subunits/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...