Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 7: 10512, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813350

ABSTRACT

It has been postulated that synaptic potentiation during waking is offset by a homoeostatic reduction in net synaptic strength during sleep. However, molecular mechanisms to support such a process are lacking. Here we demonstrate that deficiencies in the RNA-editing gene Adar increase sleep due to synaptic dysfunction in glutamatergic neurons in Drosophila. Specifically, the vesicular glutamate transporter is upregulated, leading to over-activation of NMDA receptors, and the reserve pool of glutamatergic synaptic vesicles is selectively expanded in Adar mutants. Collectively these changes lead to sustained neurotransmitter release under conditions that would otherwise result in synaptic depression. We propose that a shift in the balance from synaptic depression towards synaptic potentiation in sleep-promoting neurons underlies the increased sleep pressure of Adar-deficient animals. Our findings provide a plausible molecular mechanism linking sleep and synaptic plasticity.


Subject(s)
Adenosine Deaminase/metabolism , Drosophila Proteins/metabolism , Drosophila/enzymology , Drosophila/physiology , Glutamic Acid/metabolism , Neuronal Plasticity , Adenosine Deaminase/genetics , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Female , Male , Neurons/metabolism , Neurotransmitter Agents/metabolism , RNA Editing , Sleep , Synaptic Vesicles/metabolism
2.
Article in English | MEDLINE | ID: mdl-18419315

ABSTRACT

Sleep is controlled by two major regulatory systems: a circadian system that drives it with a 24-hour periodicity and a home-ostatic system that ensures that adequate amounts of sleep are obtained. We are using the fruit fly Drosophila melanogaster to understand both types of regulation. With respect to circadian control, we have identified molecular mechanisms that are critical for the generation of a clock. Our recent efforts have focused on the analysis of posttranslational mechanisms, specifically the action of different phosphatases that control the phosphorylation and thereby the stability and/or nuclear localization of circadian clock proteins period (PER) and timeless (TIM). Resetting the clock in response to light is also mediated through posttranslational events that target TIM for degradation by the proteasome pathway; a recently identified ubiquitin ligase, jet lag (JET), is required for this response. Our understanding of the homeostatic control of sleep is in its early stages. We have found that mushroom bodies, which are a site of synaptic plasticity in the fly brain, are important for the regulation of sleep. In addition, through analysis of genes expressed under different behavioral states, we have identified some that are up-regulated during sleep deprivation. Thus, the Drosophila model allows the use of cellular and molecular approaches that should ultimately lead to a better understanding of sleep biology.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Animals , Brain/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , F-Box Proteins/genetics , F-Box Proteins/physiology , Feedback, Physiological , Gene Expression , Genes, Insect , Models, Biological , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Period Circadian Proteins , Photoperiod , Sleep/genetics , Sleep/physiology
3.
Neuron ; 31(5): 699-711, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11567611

ABSTRACT

We describe here a general technique for the graded inhibition of cellular excitability in vivo. Inhibition is accomplished by expressing a genetically modified Shaker K(+) channel (termed the EKO channel) in targeted cells. Unlike native K(+) channels, the EKO channel strongly shunts depolarizing current: activating at potentials near E(K) and not inactivating. Selective targeting of the channel to neurons, muscles, and photoreceptors in Drosophila using the Gal4-UAS system results in physiological and behavioral effects consistent with attenuated excitability in the targeted cells, often with loss of neuronal function at higher transgene dosages. By permitting the incremental reduction of electrical activity, the EKO technique can be used to address a wide range of questions regarding neuronal function.


Subject(s)
Drosophila melanogaster/genetics , Gene Targeting , Membrane Potentials/genetics , Nervous System/metabolism , Neurons/metabolism , Potassium Channels/genetics , Adaptation, Physiological/genetics , Animals , Behavior, Animal/physiology , Cells, Cultured , Drosophila Proteins , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Female , Gene Dosage , Gene Expression Regulation, Developmental/genetics , Genes, Lethal/physiology , Larva/genetics , Larva/growth & development , Larva/metabolism , Muscles/embryology , Muscles/metabolism , Muscles/physiopathology , Mutation/physiology , Nervous System/cytology , Nervous System/embryology , Neural Inhibition/genetics , Neurons/cytology , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Phenotype , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism , Potassium Channels/metabolism , Shaker Superfamily of Potassium Channels , Synaptic Transmission/genetics , Transgenes/physiology
4.
J Biol Chem ; 276(41): 37980-5, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11495911

ABSTRACT

Calmodulin (CaM) regulates gating of several types of ion channels but has not been implicated in channel assembly or trafficking. For the SK4/IK1 K+ channel, CaM bound to the proximal C terminus ("Ct1 " domain) acts as the Ca2+ sensor. We now show that CaM interacting with the C terminus of SK4 also controls channel assembly and surface expression. In transfected cells, removing free CaM by overexpressing the CaM-binding domain, Ct1, redistributed full-length SK4 protein from the plasma membrane to the cytoplasm and decreased whole-cell currents. Making more CaM protein available by overexpressing the CaM gene abrogated the dominant-negative effect of Ct1 and restored both surface expression of SK4 protein and whole-cell currents. The distal C-terminal domain ("Ct2") also plays a role in assembly, but is not CaM-dependent. Co-immunoprecipitation experiments demonstrated that multimerization of SK4 subunits was enhanced by CaM and inhibited by removal of CaM, indicating that CaM regulates trafficking of SK4 by affecting the assembly of channels. Our results support a model in which CaM-dependent association of SK4 monomers at their Ct1 domains regulates channel assembly and surface expression. This appears to represent a novel mechanism for controlling ion channels, and consequently, the cellular functions that depend on them.


Subject(s)
Calcium/metabolism , Calmodulin/physiology , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Animals , CHO Cells , Cricetinae , Fluorescent Antibody Technique , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels , Mutagenesis , Patch-Clamp Techniques , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Binding , Protein Transport
6.
J Biol Chem ; 274(21): 14838-49, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329683

ABSTRACT

Human T lymphocytes express a Ca2+-activated K+ current (IK), whose roles and regulation are poorly understood. We amplified hSK4 cDNA from human T lymphoblasts, and we showed that its biophysical and pharmacological properties when stably expressed in Chinese hamster ovary cells were essentially identical to the native IK current. In activated lymphoblasts, hSK4 mRNA increased 14.6-fold (Kv1.3 mRNA increased 1.3-fold), with functional consequences. Proliferation was inhibited when Kv1.3 and IK were blocked in naive T cells, but IK block alone inhibited re-stimulated lymphoblasts. IK and Kv1.3 were involved in volume regulation, but IK was more important, particularly in lymphoblasts. hSK4 lacks known Ca2+-binding sites; however, we mapped a Ca2+-dependent calmodulin (CaM)-binding site to the proximal C terminus (Ct1) of hSK4. Full-length hSK4 produced a highly negative membrane potential (Vm) in Chinese hamster ovary cells, whereas the channels did not function when either Ct1 or the distal C terminus was deleted (Vm approximately 0 mV). Native IK (but not expressed hSK4) current was inhibited by CaM and CaM kinase antagonists at physiological Vm values, suggesting modulation by an accessory molecule in native cells. Our results provide evidence for increased roles for IK/hSK4 in activated T cell functions; thus hSK4 may be a promising therapeutic target for disorders involving the secondary immune response.


Subject(s)
Potassium Channels, Calcium-Activated , Potassium Channels, Voltage-Gated , Potassium Channels/physiology , T-Lymphocytes/cytology , Animals , CHO Cells , Calmodulin/metabolism , Cell Division , Cricetinae , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels , Kv1.3 Potassium Channel , Membrane Potentials , Potassium Channels/genetics , RNA, Messenger/biosynthesis , Up-Regulation
7.
Nat Neurosci ; 1(6): 462-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-10196543

ABSTRACT

Large-conductance calcium-activated potassium channels (maxi-K channels) have an essential role in the control of excitability and secretion. Only one gene Slo is known to encode maxi-K channels, which are sensitive to both membrane potential and intracellular calcium. We have isolated a potassium channel gene called Slack that is abundantly expressed in the nervous system. Slack channels rectify outwardly with a unitary conductance of about 25-65 pS and are inhibited by intracellular calcium. However, when Slack is co-expressed with Slo, channels with pharmacological properties and single-channel conductances that do not match either Slack or Slo are formed. The Slack/Slo channels have intermediate conductances of about 60-180 pS and are activated by cytoplasmic calcium. Our findings indicate that some intermediate-conductance channels in the nervous system may result from an interaction between Slack and Slo channel subunits.


Subject(s)
Nerve Tissue Proteins , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Potassium Channels/physiology , Amino Acid Sequence/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins , Electric Conductivity , Intermediate-Conductance Calcium-Activated Potassium Channels , Isomerism , Large-Conductance Calcium-Activated Potassium Channels , Molecular Sequence Data , Potassium Channels/genetics , Potassium Channels, Sodium-Activated
8.
Proc Natl Acad Sci U S A ; 94(20): 11013-8, 1997 Sep 30.
Article in English | MEDLINE | ID: mdl-9380751

ABSTRACT

The gene for hSK4, a novel human small conductance calcium-activated potassium channel, or SK channel, has been identified and expressed in Chinese hamster ovary cells. In physiological saline hSK4 generates a conductance of approximately 12 pS, a value in close agreement with that of other cloned SK channels. Like other members of this family, the polypeptide encoded by hSK4 contains a previously unnoted leucine zipper-like domain in its C terminus of unknown function. hSK4 appears unique, however, in its very high affinity for Ca2+ (EC50 of 95 nM) and its predominant expression in nonexcitable tissues of adult animals. Together with the relatively low homology of hSK4 to other SK channel polypeptides (approximately 40% identical), these data suggest that hSK4 belongs to a novel subfamily of SK channels.


Subject(s)
Calcium/metabolism , Potassium Channels, Calcium-Activated , Potassium Channels/genetics , Amino Acid Sequence , Animals , Barium/metabolism , CHO Cells , Cloning, Molecular , Cricetinae , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels , Molecular Sequence Data , Potassium Channels/metabolism , Sequence Homology, Amino Acid
9.
Nature ; 376(6542): 690-5, 1995 Aug 24.
Article in English | MEDLINE | ID: mdl-7651518

ABSTRACT

Potassium channels catalyse the permeation of K+ ions across cellular membranes and are identified by a common structural motif, a highly conserved signature sequence of eight amino acids in the P domain of each channel's pore-forming alpha-subunit. Here we describe a novel K+ channel (TOK1) from Saccharomyces cerevisiae that contains two P domains within one continuous polypeptide. Xenopus laevis oocytes expressing the channel exhibit a unique, outwardly rectifying, K(+)-selective current. The channel is permeable to outward flow of ions at membrane potentials above the K+ equilibrium potential; its conduction-voltage relationship is thus sensitive to extracellular K+ ion concentration. In excised membrane patches, external divalent cations block the channel in a voltage-dependent manner, and their removal in this configuration allows inward channel current. These attributes are similar to those described for inwardly rectifying K+ channels, but in the opposite direction, a previously unrecognized channel behaviour. Our results identify a new class of K+ channel which is distinctive in both its primary structure and functional properties. Structural homologues of the channel are present in the genome of Caenorhabditis elegans.


Subject(s)
Potassium Channels/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans , Cells, Cultured , DNA Primers , Drosophila , Molecular Sequence Data , Oocytes , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Conformation , Recombinant Proteins , Sequence Homology, Amino Acid , Sodium/metabolism , Xenopus laevis
10.
Neuron ; 12(2): 301-15, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8110460

ABSTRACT

In response to changes in membrane potential, voltage-dependent ion channel proteins undergo conformational rearrangements that lead to channel opening. These rearrangements move a net charge, measured as "gating current", across the membrane. Here we characterize the effects of the pharmacological blocker 4-aminopyridine on both the K+ and gating currents of wild-type and mutant Shaker K+ channels. Our results indicate that the activation of these channels involves two distinct types of structural rearrangement. In addition to independent Hodgkin and Huxley type rearrangements for each of the four subunits, which are responsible for most of the gating charge movement, Shaker channels interconvert between two quaternary conformations during activation. The transition between the two quaternary states moves about 10% of the total gating charge, and it is selectively blocked by 4-aminopyridine.


Subject(s)
Gene Rearrangement , Mutation , Potassium Channels/chemistry , Potassium Channels/physiology , 4-Aminopyridine/metabolism , 4-Aminopyridine/pharmacology , Animals , Drosophila , Electrophysiology , Ion Channel Gating/drug effects , Mathematics , Molecular Conformation , Potassium Channels/genetics , Potassium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...