Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 118(5): 523-46, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696610

ABSTRACT

CNG channels in vivo are heteromers of homologous alpha and beta subunits that each contain a six-transmembrane segment domain and a COOH-terminal cytoplasmic cyclic nucleotide binding domain (BD). In heterologous expression systems, heteromeric alphabeta channels activate with greater sensitivity to ligand than do homomeric alpha channels; however, ligand-gating of channels containing only beta subunit BDs has never been studied because beta subunits cannot form functional homomeric CNG channels. To characterize directly the contribution of the beta subunit BD to ligand-gating, we constructed a chimeric subunit, X-beta, whose BD sequence was that of the beta subunit CNG5 from rat, but whose sequence outside the BD was derived from alpha subunits. For comparison, we constructed another chimera, X-alpha, whose sequence outside the BD was identical to that of X-beta, but whose BD sequence was that of the alpha subunit CNG2 from catfish. When expressed in Xenopus oocytes, X-beta and X-alpha each formed functional homomeric channels activated by both cAMP and cGMP. This is the first demonstration that the beta subunit BD can couple ligand binding to activation in the absence of alpha subunit BD residues. Notably, both agonists activate X-beta more effectively than X-alpha (higher opening efficacy and lower K(1/2)). The BD is believed to comprise two functionally distinct subdomains: (1) the roll subdomain (beta-roll and flanking A- and B-helices) and (2) the C-helix subdomain. Opening efficacy was previously believed to be controlled primarily by the C-helix, but when we made additional chimeras by exchanging the subdomains between X-beta and X-alpha, we found that both subdomains contain significant determinants of efficacy and agonist selectivity. In particular, only channels containing the roll subdomain of the beta subunit had high efficacy. Thermodynamic linkage analysis shows that interaction between the two subdomains accounts for a significant portion of their contribution to activation energetics.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Ion Channels/physiology , Animals , Binding Sites/physiology , Chimera , Isomerism , Ligands , Oocytes , Rats , Signal Transduction , Thermodynamics , Xenopus
2.
J Gen Physiol ; 118(3): 237-50, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524455

ABSTRACT

Members of the hyperpolarization-activated cation (HCN) channel family generate HCN currents (I(h)) that are directly regulated by cAMP and contribute to pacemaking activity in heart and brain. The four different HCN isoforms show distinct biophysical properties. In cell-free patches from Xenopus oocytes, the steady-state activation curve of HCN2 channels is 20 mV more hyperpolarized compared with HCN1. Whereas the binding of cAMP to a COOH-terminal cyclic nucleotide binding domain (CNBD) markedly shifts the activation curve of HCN2 by 17 mV to more positive potentials, the response of HCN1 is much less pronounced (4 mV shift). A previous deletion mutant study suggested that the CNBD inhibits hyperpolarization-gating in the absence of cAMP; the binding of cAMP shifts gating to more positive voltages by relieving this inhibition. The differences in basal gating and cAMP responsiveness between HCN1 and HCN2 were proposed to result from a greater inhibitory effect of the CNBD in HCN2 compared with HCN1. Here, we use a series of chimeras between HCN1 and HCN2, in which we exchange the NH(2) terminus, the transmembrane domain, or distinct domains of the COOH terminus, to investigate further the molecular bases for the modulatory action of cAMP and for the differences in the functional properties of the two channels. Differences in cAMP regulation between HCN1 and HCN2 are localized to sequence differences within the COOH terminus of the two channels. Surprisingly, exchange of the CNBDs between HCN1 and HCN2 has little effect on basal gating and has only a modest one on cAMP modulation. Rather, differences in cAMP modulation depend on the interaction between the CNBD and the C-linker, a conserved 80-amino acid region that connects the last (S6) transmembrane segment to the CNBD. Differences in basal gating depend on both the core transmembrane domain and the COOH terminus. These data, taken in the context of the previous data on deletion mutants, suggest that the inhibitory effect of the CNBD on basal gating depends on its interactions with both the C-linker and core transmembrane domain of the channel. The extent to which cAMP binding is able to relieve this inhibition is dependent on the interaction between the C-linker and the CNBD.


Subject(s)
Cyclic AMP/physiology , Ion Channel Gating/physiology , Ion Channels/physiology , Nerve Tissue Proteins , Animals , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Membranes/metabolism , Mice , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels , Xenopus
3.
Nature ; 411(6839): 805-10, 2001 Jun 14.
Article in English | MEDLINE | ID: mdl-11459060

ABSTRACT

Hyperpolarization-activated cation channels of the HCN gene family contribute to spontaneous rhythmic activity in both heart and brain. All four family members contain both a core transmembrane segment domain, homologous to the S1-S6 regions of voltage-gated K+ channels, and a carboxy-terminal 120 amino-acid cyclic nucleotide-binding domain (CNBD) motif. Homologous CNBDs are responsible for the direct activation of cyclic nucleotide-gated channels and for modulation of the HERG voltage-gated K+ channel--important for visual and olfactory signalling and for cardiac repolarization, respectively. The direct binding of cyclic AMP to the cytoplasmic site on HCN channels permits the channels to open more rapidly and completely after repolarization of the action potential, thereby accelerating rhythmogenesis. However, the mechanism by which cAMP binding modulates HCN channel gating and the basis for functional differences between HCN isoforms remain unknown. Here we demonstrate by constructing truncation mutants that the CNBD inhibits activation of the core transmembrane domain. cAMP binding relieves this inhibition. Differences in activation gating and extent of cAMP modulation between the HCN1 and HCN2 isoforms result largely from differences in the efficacy of CNBD inhibition.


Subject(s)
Cyclic AMP/metabolism , Ion Channels/metabolism , Muscle Proteins , Nerve Tissue Proteins , Animals , Binding Sites , Cell Membrane/metabolism , Cloning, Molecular , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating , Mice , Models, Molecular , Mutagenesis , Potassium Channels , Protein Conformation
4.
Nat Neurosci ; 4(7): 711-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11426227

ABSTRACT

Controversy exists regarding the site of modification of synaptic transmission during long-term plasticity in the mammalian hippocampus. Here we used a fluorescent marker of presynaptic activity, FM 1-43, to directly image changes in presynaptic function during both short-term and long-term forms of plasticity at presynaptic boutons of CA3-CA1 excitatory synapses in acute hippocampal slices. We demonstrated enhanced presynaptic function during long-term potentiation (LTP) induced either chemically (with tetraethylammonium), or by high-frequency (200-Hz) electrical stimulation. Both of these forms of LTP required activation of L-type voltage-gated calcium channels and NMDA receptors in the postsynaptic CA1 neuron. These results thus implied that a long-lasting increase in the efficacy of synaptic transmission is likely to depend, at least in part, on enhanced transmitter release from the presynaptic neuron.


Subject(s)
Neuronal Plasticity , Presynaptic Terminals/physiology , Animals , Electric Stimulation , Excitatory Postsynaptic Potentials , Fluorescent Dyes , Hippocampus/physiology , Hippocampus/ultrastructure , In Vitro Techniques , Long-Term Potentiation , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Pyridinium Compounds , Quaternary Ammonium Compounds
5.
J Gen Physiol ; 117(5): 491-504, 2001 May.
Article in English | MEDLINE | ID: mdl-11331358

ABSTRACT

Members of the HCN channel family generate hyperpolarization-activated cation currents (Ih) that are directly regulated by cAMP and contribute to pacemaker activity in heart and brain. The four HCN isoforms show distinct but overlapping patterns of expression in different tissues. Here, we report that HCN1 and HCN2, isoforms coexpressed in neocortex and hippocampus that differ markedly in their biophysical properties, coassemble to generate heteromultimeric channels with novel properties. When expressed in Xenopus oocytes, HCN1 channels activate 5-10-fold more rapidly than HCN2 channels. HCN1 channels also activate at voltages that are 10-20 mV more positive than those required to activate HCN2. In cell-free patches, the steady-state activation curve of HCN1 channels shows a minimal shift in response to cAMP (+4 mV), whereas that of HCN2 channels shows a pronounced shift (+17 mV). Coexpression of HCN1 and HCN2 yields Ih currents that activate with kinetics and a voltage dependence that tend to be intermediate between those of HCN1 and HCN2 homomers, although the coexpressed channels do show a relatively large shift by cAMP (+14 mV). Neither the kinetics, steady-state voltage dependence, nor cAMP dose-response curve for the coexpressed Ih can be reproduced by the linear sum of independent populations of HCN1 and HCN2 homomers. These results are most simply explained by the formation of heteromeric channels with novel properties. The properties of these heteromeric channels closely resemble the properties of I(h) in hippocampal CA1 pyramidal neurons, cells that coexpress HCN1 and HCN2. Finally, differences in Ih channel properties recorded in cell-free patches versus intact oocytes are shown to be due, in part, to modulation of Ih by basal levels of cAMP in intact cells.


Subject(s)
Biological Clocks/physiology , Cyclic AMP/metabolism , Ion Channel Gating/physiology , Ion Channels/genetics , Ion Channels/metabolism , Muscle Proteins , Nerve Tissue Proteins , Animals , Binding Sites/physiology , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels , Dose-Response Relationship, Drug , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating/drug effects , Ion Channels/chemistry , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mutagenesis, Site-Directed/physiology , Oocytes/physiology , Patch-Clamp Techniques , Point Mutation/physiology , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , Xenopus
6.
Nat Neurosci ; 3(11): 1107-12, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11036267

ABSTRACT

Although the function of the p42/p44 mitogen-activated protein (MAP) kinase pathway in long-term potentiation at hippocampal CA3-CA1 synapses has been well described, relatively little is known about the importance of the p38 MAP kinase pathway in synaptic plasticity. Here we show that the p38 MAP kinase pathway, a parallel signaling cascade activated by distinct upstream kinases, mediates the induction of metabotropic glutamate receptor-dependent long-term depression at CA3-CA1 synapses. Thus, two parallel MAP kinase pathways contribute to opposing forms of long-term plasticity at a central synapse.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Electric Stimulation , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Hippocampus/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Synapses/drug effects , p38 Mitogen-Activated Protein Kinases
7.
J Neurosci ; 20(14): 5264-75, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10884310

ABSTRACT

The hyperpolarization-activated cation current (termed I(h), I(q), or I(f)) was recently shown to be encoded by a new family of genes, named HCN for hyperpolarization-activated cyclic nucleotide-sensitive cation nonselective. When expressed in heterologous cells, each HCN isoform generates channels with distinct activation kinetics, mirroring the range of biophysical properties of native I(h) currents recorded in different classes of neurons. To determine whether the functional diversity of I(h) currents is attributable to different patterns of HCN gene expression, we determined the mRNA distribution across different regions of the mouse CNS of the three mouse HCN genes that are prominently expressed there (mHCN1, 2 and 4). We observe distinct patterns of distribution for each of the three genes. Whereas mHCN2 shows a widespread expression throughout the CNS, the expression of mHCN1 and mHCN4 is more limited, and generally complementary. mHCN1 is primarily expressed within neurons of the neocortex, hippocampus, and cerebellar cortex, but also in selected nuclei of the brainstem. mHCN4 is most highly expressed within neurons of the medial habenula, thalamus, and olfactory bulb, but also in distinct neuronal populations of the basal ganglia. Based on a comparison of mRNA expression with an electrophysiological characterization of native I(h) currents in hippocampal and thalamic neurons, our data support the idea that the functional heterogeneity of I(h) channels is attributable, in part, to differential isoform expression. Moreover, in some neurons, specific functional roles can be proposed for I(h) channels with defined subunit composition.


Subject(s)
Biological Clocks/physiology , Central Nervous System/metabolism , Ion Channels/metabolism , Muscle Proteins , Nerve Tissue Proteins , Animals , Biological Clocks/genetics , Brain/metabolism , Cells, Cultured , Central Nervous System/cytology , Cyclic Nucleotide-Gated Cation Channels , Gene Expression , Hippocampus/cytology , Hippocampus/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Vitro Techniques , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Multigene Family , Neurons/cytology , Neurons/metabolism , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spinal Cord/metabolism , Thalamus/cytology , Thalamus/metabolism , Xenopus
9.
Neuron ; 28(3): 899-909, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11163275

ABSTRACT

The structure of the pore region of the alpha subunit of the bovine rod cyclic nucleotide-gated channel was probed using cysteine-scanning mutagenesis and hydrophilic sulfhydryl-reactive methanethiosulfonate (MTS) reagents. A region homologous to the pore helix in the X-ray crystal structure of the KcsA K(+) channel showed a helical pattern of reactivity with externally applied MTS reagents. Surprisingly, three out of four of the reactive residues, all on one face of the pore helix, only reacted with MTS reagents in the closed state. A residue on the opposite face of the helix only reacted with MTS reagents in the open state. These results indicate that the pore helix (or its surroundings) undergoes a change in conformation, perhaps involving a rotation around its long axis, that opens a gate localized to the selectivity filter of the channel.


Subject(s)
Bacterial Proteins , Ion Channel Gating/physiology , Ion Channels/genetics , Ion Channels/metabolism , Animals , Cattle , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Indicators and Reagents , Ion Channel Gating/drug effects , Mesylates/pharmacology , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Conformation/drug effects , Protein Structure, Secondary/drug effects , Protein Structure, Secondary/physiology , Retinal Rod Photoreceptor Cells/metabolism , Sequence Homology, Amino Acid , Sulfhydryl Reagents/pharmacology , Water/metabolism , Xenopus
10.
Neuron ; 24(3): 687-700, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10595519

ABSTRACT

We have generated mice lacking synaptogyrin I and synaptophysin I to explore the functions of these abundant tyrosine-phosphorylated proteins of synaptic vesicles. Single and double knockout mice were alive and fertile without significant morphological or biochemical changes. Electrophysiological recordings in the hippocampal CA1 region revealed that short-term and long-term synaptic plasticity were severely reduced in the synaptophysin/synaptogyrin double knockout mice. LTP was decreased independent of the induction protocol, suggesting that the defect in LTP was not caused by insufficient induction. Our data show that synaptogyrin I and synaptophysin I perform redundant and essential functions in synaptic plasticity without being required for neurotransmitter release itself.


Subject(s)
Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Synaptophysin/physiology , Animals , Brain/pathology , Electric Stimulation , Long-Term Potentiation/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout/genetics , Mice, Knockout/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurotransmitter Agents/metabolism , Pedigree , Synaptogyrins , Synaptophysin/deficiency , Synaptophysin/genetics , Time Factors
12.
Annu Rev Neurosci ; 22: 443-85, 1999.
Article in English | MEDLINE | ID: mdl-10202545

ABSTRACT

The quantity of neurotransmitter released into the synaptic cleft, the reliability with which it is released, and the response of the postsynaptic cell to that transmitter all contribute to the strength of a synaptic connection. The presynaptic nerve terminal is a major regulatory site for activity-dependent changes in synaptic function. Ionotropic receptors for the inhibitory amino acid GABA, expressed on the presynaptic terminals of crustacean motor axons and vertebrate sensory neurons, were the first well-defined mechanism for the heterosynaptic transmitter-mediated regulation of transmitter release. Recently, presynaptic ionotropic receptors for a large range of transmitters have been found to be widespread throughout the central and peripheral nervous systems. In this review, we first consider some general theoretical issues regarding whether and how presynaptic ionotropic receptors are important regulators of presynaptic function. We consider the criteria that should be met to identify a presynaptic ionotropic receptor and its regulatory function and review several examples of presynaptic receptors that meet at least some of those criteria. We summarize the classic studies of presynaptic inhibition mediated by GABA-gated Cl channels and then focus on presynaptic nicotinic ACh receptors and presynaptic glutamate receptors. Finally, we briefly discuss evidence for other types of presynaptic ionotropic receptors.


Subject(s)
Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Receptors, Cell Surface/physiology , Acetylcholine/physiology , Animals , Humans , Ion Channel Gating/physiology , Ion Channels/physiology , Neural Inhibition/physiology , Receptors, Cell Surface/metabolism , Receptors, GABA-A/physiology , Receptors, Glutamate/physiology
13.
Nat Neurosci ; 2(1): 24-30, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10195176

ABSTRACT

Long-term forms of synaptic plasticity that may underlie learning and memory have been suggested to depend on changes in the number of synapses between presynaptic and postsynaptic neurons. Here we have investigated a form of synaptic plasticity in cultures of hippocampal CA3 and CA1 neurons related to the late phase of long-term potentiation, which depends on cAMP and protein synthesis. Using the fluorescent dye FM 1-43 to label active presynaptic terminals, we find that a membrane permeable analog of cAMP enhances the number of active presynaptic terminals and that this effect requires protein synthesis.


Subject(s)
Cyclic AMP/physiology , Hippocampus/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Animals , Anisomycin/pharmacology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Fluorescent Dyes , Hippocampus/cytology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Presynaptic Terminals/drug effects , Protein Synthesis Inhibitors/pharmacology , Pyridinium Compounds , Quaternary Ammonium Compounds , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/physiology , Thionucleotides/pharmacology
14.
J Gen Physiol ; 113(1): 17-34, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9874685

ABSTRACT

Cyclic nucleotide-gated channels are composed of a core transmembrane domain, structurally homologous to the voltage-gated K+ channels, and a cytoplasmic ligand-binding domain. These two modules are joined by approximately 90 conserved amino acids, the C-linker, whose precise role in the mechanism of channel activation by cyclic nucleotides is poorly understood. We examined cyclic nucleotide-gated channels from bovine photoreceptors and Caenorhabditis elegans sensory neurons that show marked differences in cyclic nucleotide efficacy and sensitivity. By constructing chimeras from these two channels, we identified a region of 30 amino acids in the C-linker (the L2 region) as an important determinant of activation properties. An increase in both the efficacy of gating and apparent affinity for cGMP and cAMP can be conferred onto the photoreceptor channel by the replacement of its L2 region with that of the C. elegans channel. Three residues within this region largely account for this effect. Despite the profound effect of the C-linker region on ligand gating, the identity of the C-linker does not affect the spontaneous, ligand-independent open probability. Based on a cyclic allosteric model of activation, we propose that the C-linker couples the opening reaction in the transmembrane core region to the enhancement of the affinity of the open channel for agonist, which underlies ligand gating.


Subject(s)
Ion Channel Gating/genetics , Ion Channel Gating/physiology , Mutagenesis, Insertional , Nucleotides, Cyclic/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans , Cattle , Electric Stimulation , Electrophysiology , Ligands , Membrane Potentials/physiology , Molecular Sequence Data , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Patch-Clamp Techniques , Photoreceptor Cells, Vertebrate/physiology , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels/physiology
15.
J Neurosci ; 18(21): 8770-9, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9786984

ABSTRACT

The cellular mechanisms underlying the induction and expression of homosynaptic depression at the glutamatergic synapse between Aplysia sensory and motor neurons were studied in dissociated cell culture. Intracellular microelectrodes were used to stimulate action potentials in the presynaptic sensory neuron and record the depolarizing EPSP from the motor neuron. Homosynaptic depression (HSD) was induced by repeatedly stimulating the sensory neuron at rates as low as one action potential per minute. Activation of postsynaptic Glu receptors was neither sufficient nor necessary to induce HSD. Thus, repeated applications of exogenous Glu did not depress the synaptically evoked EPSP. Moreover, normal HSD was observed when the sensory neuron was stimulated during a period when the Glu receptors were blocked with the antagonist DNQX. The induction of HSD is thus likely to occur within the presynaptic terminal. We explored the role of presynaptic calcium in the induction of HSD by injecting the sensory neuron with EGTA, a relatively slow calcium chelator that does not alter rapid release but effectively buffers the slow residual calcium transient thought to be important for plasticity. EGTA had little effect on HSD, indicating that residual Cai is not involved. HSD does not appear to involve a decrease in presynaptic calcium influx, because there was no change in the presynaptic calcium transient, measured by calcium indicator dyes, during HSD. We conclude that HSD is induced and expressed in the presynaptic terminal, possibly by a mechanism directly coupled to the release process.


Subject(s)
Motor Neurons/physiology , Neuronal Plasticity/physiology , Neurons, Afferent/physiology , Presynaptic Terminals/physiology , Synaptic Transmission/physiology , Animals , Aplysia , Calcium/physiology , Cells, Cultured , Egtazic Acid/pharmacology , Evoked Potentials/drug effects , Motor Neurons/drug effects , Neuronal Plasticity/drug effects , Neurons, Afferent/drug effects , Synaptic Transmission/drug effects
16.
Neuron ; 21(1): 235-48, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9697867

ABSTRACT

Cyclic nucleotide-gated ion channels are composed of four pore-forming subunits. Binding of cyclic nucleotide to a site in the intracellular carboxyl terminus of each subunit leads to channel activation. Since there are four subunits, four binding events are possible. In this study, we investigate the effects of individual binding events on activation by studying channels containing one, two, three, or four functional binding sites. The binding of a single ligand significantly increases opening, although four ligands are required for full activation. The data are inconsistent with models in which the four subunits activate in a single concerted step (Monod-Wyman-Changeux model) or in four independent steps (Hodgkin-Huxley model). Instead, the four subunits may associate and activate as two independent dimers.


Subject(s)
Ion Channel Gating/physiology , Ion Channels/physiology , Models, Biological , Nucleotides, Cyclic/physiology , Animals , Binding Sites/physiology , Cattle , Chemical Phenomena , Chemistry , Dimerization , Ion Channels/chemistry , Ion Channels/genetics , Ligands , Mathematics , Point Mutation
17.
Cell ; 93(5): 717-29, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9630217

ABSTRACT

The generation of pacemaker activity in heart and brain is mediated by hyperpolarization-activated cation channels that are directly regulated by cyclic nucleotides. We previously cloned a novel member of the voltage-gated K channel family from mouse brain (mBCNG-1) that contained a carboxy-terminal cyclic nucleotide-binding domain (Santoro et al., 1997) and hence proposed it to be a candidate gene for pacemaker channels. Heterologous expression of mBCNG-1 demonstrates that it does indeed code for a channel with properties indistinguishable from pacemaker channels in brain and similar to those in heart. Three additional mouse genes and two human genes closely related to mBCNG-1 display unique patterns of mRNA expression in different tissues, including brain and heart, demonstrating that these channels constitute a widely expressed gene family.


Subject(s)
Biological Clocks/genetics , Brain/physiology , Ion Channels/genetics , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Animals , Barium/pharmacology , Cesium/pharmacology , Cloning, Molecular , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels , DNA, Complementary/genetics , Electric Conductivity , Gene Expression , Heart/physiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/antagonists & inhibitors , Ion Channels/biosynthesis , Mice , Molecular Sequence Data , Multigene Family , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Neuroglia/metabolism , Oocytes , Pacemaker, Artificial , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels , Recombinant Proteins/biosynthesis , Sequence Homology, Amino Acid , Sodium/metabolism , Species Specificity , Tissue Distribution , Xenopus
18.
J Neurophysiol ; 79(2): 870-8, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9463448

ABSTRACT

The effects of prostaglandin E2 (PGE2), an important metabolite of arachidonic acid, were studied on the activity of nicotinic AChR channels in cultured chick sympathetic ganglion neurons. In whole cell recordings, PGE2 (25 nM) inhibited significantly the ACh-evoked macroscopic current. In cell-attached patch recordings, PGE2 significantly inhibited single AChR channel currents as a result of a decrease in the frequency of channel opening, with no change in open time and conductance. PGE2 did not alter the extent or rate of agonist-induced desensitization of the AChR channels. These effects are specific since the related compound PGD2 had no effect on AChR channel function. Because there is an abundant endogenous production of PGE2 within sympathetic ganglia in response to certain stimuli, the inhibition of AChR channel function by PGE2 could serve an important role to modulate synaptic transmission in the sympathetic nervous system.


Subject(s)
Dinoprostone/pharmacology , Ganglia, Sympathetic/cytology , Neurons/drug effects , Receptors, Nicotinic/drug effects , Animals , Calcium/physiology , Chick Embryo , Cyclooxygenase Inhibitors/pharmacology , Ganglia, Sympathetic/physiology , Indomethacin/pharmacology , Ion Channel Gating/drug effects , Neurons/physiology , Norepinephrine/pharmacology , Patch-Clamp Techniques , Prostaglandin D2/pharmacology , Receptors, Nicotinic/physiology , Synaptic Transmission
19.
J Biol Chem ; 273(8): 4497-505, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9468504

ABSTRACT

Activation of cyclic nucleotide-gated channels is thought to involve two distinct steps: a recognition event in which a ligand binds to the channel and a conformational change that both opens the channel and increases the affinity of the channel for an agonist. Sequence similarity with the cyclic nucleotide-binding sites of cAMP- and cGMP-dependent protein kinases and the bacterial catabolite activating protein (CAP) suggests that the channel ligand binding site consists of a beta-roll and three alpha-helices. Recent evidence has demonstrated that the third (or C) alpha-helix moves relative to the agonist upon channel activation, forming additional favorable contacts with the purine ring. Here we ask if channel activation also involves structural changes in the beta-roll by investigating the contribution of a conserved arginine residue that, in CAP and the kinases, forms an important ionic interaction with the cyclized phosphate of the bound ligand. Mutations that conserve, neutralize, or reverse the charge on this arginine decreased the apparent affinity for ligand over four orders of magnitude but had little effect on the ability of bound ligand to open the channel. These data indicate that the cyclized phosphate of the nucleotide approaches to within 2-4 A of the arginine, forming a favorable ionic bond that is largely unaltered upon activation. Thus, the binding site appears to be polarized into two distinct structural and functional domains: the beta-roll stabilizes the ligand in a state-independent manner, whereas the C-helix selectively stabilizes the ligand in the open state of the channel. It is likely that these distinct contributions of the nucleotide/C-helix and nucleotide/beta-roll interactions may also be a general feature of the mechanism of activation of other cyclic nucleotide-binding proteins.


Subject(s)
Arginine/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Ion Channel Gating , Ion Channels/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Binding Sites , Conserved Sequence , Ion Channels/chemistry , Ion Channels/physiology , Molecular Sequence Data , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Xenopus
20.
Neuron ; 19(3): 635-51, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9331354

ABSTRACT

Long-term potentiation at CA3-CA1 hippocampal synapses exhibits an early phase and a late phase, which can be distinguished by their underlying molecular mechanisms. Unlike the early phase, the late phase is dependent on both cAMP and protein synthesis. Quantal analysis of unitary synaptic transmission between a single presynaptic CA3 neuron and a single postsynaptic CA1 neuron suggests that, under certain conditions, the early phase of LTP involves an increase in the probability of release of a single quantum of transmitter from a single presynaptic release site, with no change in the number of quanta that are released or in postsynaptic sensitivity to transmitter. Here, we show that the cAMP-induced late phase of LTP involves an increase in the number of quanta released in response to a single presynaptic action potential, possibly due to an increase in the number of sites of synaptic transmission between a single CA3 and a single CA1 neuron.


Subject(s)
Cyclic AMP/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Synaptic Transmission/physiology , Animals , Calcium/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Excitatory Postsynaptic Potentials/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/metabolism , Synaptic Membranes/chemistry , Synaptic Membranes/physiology , Synaptic Transmission/drug effects , Thionucleotides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...