Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 16099, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30382129

ABSTRACT

Functional synaptic networks are compromised in many neurodevelopmental and neurodegenerative diseases. While the mechanisms of axonal transport and localization of synaptic vesicles and mitochondria are relatively well studied, little is known about the mechanisms that regulate the localization of proteins that localize to active zones. Recent finding suggests that mechanisms involved in transporting proteins destined to active zones are distinct from those that transport synaptic vesicles or mitochondria. Here we report that localization of BRP-an essential active zone scaffolding protein in Drosophila, depends on the precise balance of neuronal Par-1 kinase. Disruption of Par-1 levels leads to excess accumulation of BRP in axons at the expense of BRP at active zones. Temporal analyses demonstrate that accumulation of BRP within axons precedes the loss of synaptic function and its depletion from the active zones. Mechanistically, we find that Par-1 co-localizes with BRP and is present in the same molecular complex, raising the possibility of a novel mechanism for selective localization of BRP-like active zone scaffolding proteins. Taken together, these data suggest an intriguing possibility that mislocalization of active zone proteins like BRP might be one of the earliest signs of synapse perturbation and perhaps, synaptic networks that precede many neurological disorders.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glycogen Synthase Kinase 3/metabolism , Neuromuscular Junction/metabolism , Synapses/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Larva/metabolism , Larva/ultrastructure , Microtubule-Associated Proteins/metabolism , Presynaptic Terminals/metabolism , Protein Transport , Synapses/ultrastructure
3.
Development ; 141(23): 4548-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359729

ABSTRACT

Synaptic scaffold proteins control the localization of ion channels and receptors, and facilitate molecular associations between signaling components that modulate synaptic transmission and plasticity. Here, we define novel roles for a recently described scaffold protein, Dsychronic (DYSC), at the Drosophila larval neuromuscular junction. DYSC is the Drosophila homolog of whirlin/DFNB31, a PDZ domain protein linked to Usher syndrome, the most common form of human deaf-blindness. We show that DYSC is expressed presynaptically and is often localized adjacent to the active zone, the site of neurotransmitter release. Loss of DYSC results in marked alterations in synaptic morphology and cytoskeletal organization. Moreover, active zones are frequently enlarged and misshapen in dysc mutants. Electrophysiological analyses further demonstrate that dysc mutants exhibit substantial increases in both evoked and spontaneous synaptic transmission. We have previously shown that DYSC binds to and regulates the expression of the Slowpoke (SLO) BK potassium channel. Consistent with this, slo mutant larvae exhibit similar alterations in synapse morphology, active zone size and neurotransmission, and simultaneous loss of dysc and slo does not enhance these phenotypes, suggesting that dysc and slo act in a common genetic pathway to modulate synaptic development and output. Our data expand our understanding of the neuronal functions of DYSC and uncover non-canonical roles for the SLO potassium channel at Drosophila synapses.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Proteins/metabolism , Neuromuscular Junction/growth & development , Synapses/physiology , Animals , Immunohistochemistry , Larva/growth & development , Membrane Potentials , Microscopy, Confocal , PDZ Domains/genetics , Patch-Clamp Techniques , Reverse Transcriptase Polymerase Chain Reaction , Synapses/metabolism
4.
J Neurosci ; 33(50): 19590-8, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24336723

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease that leads invariably to fatal paralysis associated with motor neuron degeneration and muscular atrophy. One gene associated with ALS encodes the DNA/RNA-binding protein Fused in Sarcoma (FUS). There now exist two Drosophila models of ALS. In one, human FUS with ALS-causing mutations is expressed in fly motor neurons; in the other, the gene cabeza (caz), the fly homolog of FUS, is ablated. These FUS-ALS flies exhibit larval locomotor defects indicative of neuromuscular dysfunction and early death. The locus and site of initiation of this neuromuscular dysfunction remain unclear. We show here that in FUS-ALS flies, motor neuron cell bodies fire action potentials that propagate along the axon and voltage-dependent inward and outward currents in the cell bodies are indistinguishable in wild-type and FUS-ALS motor neurons. In marked contrast, the amplitude of synaptic currents evoked in the postsynaptic muscle cell is decreased by >80% in FUS-ALS larvae. Furthermore, the frequency but not unitary amplitude of spontaneous miniature synaptic currents is decreased dramatically in FUS-ALS flies, consistent with a change in quantal content but not quantal size. Although standard confocal microscopic analysis of the larval neuromuscular junction reveals no gross abnormalities, superresolution stimulated emission depletion (STED) microscopy demonstrates that the presynaptic active zone protein bruchpilot is aberrantly organized in FUS-ALS larvae. The results are consistent with the idea that defects in presynaptic terminal structure and function precede, and may contribute to, the later motor neuron degeneration that is characteristic of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Nerve Degeneration/pathology , RNA-Binding Protein FUS/metabolism , Synapses/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Drosophila , Motor Neurons/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , RNA-Binding Protein FUS/genetics , Synapses/genetics , Synapses/metabolism
5.
J Neurosci ; 33(42): 16767-77, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133277

ABSTRACT

SLOB (SLOWPOKE-binding protein) modulates the Drosophila SLOWPOKE calcium-activated potassium channel. We have shown previously that SLOB deletion or RNAi knockdown decreases excitability of neurosecretory pars intercerebralis (PI) neurons in the adult Drosophila brain. In contrast, we found that SLOB deletion/knockdown enhances neurotransmitter release from motor neurons at the fly larval neuromuscular junction, suggesting an increase in excitability. Because two prominent SLOB isoforms, SLOB57 and SLOB71, modulate SLOWPOKE channels in opposite directions in vitro, we investigated whether divergent expression patterns of these two isoforms might underlie the differential modulation of excitability in PI and motor neurons. By performing detailed in vitro and in vivo analysis, we found strikingly different modes of regulatory control by the slob57 and slob71 promoters. The slob71, but not slob57, promoter contains binding sites for the Hunchback and Mirror transcriptional repressors. Furthermore, several core promoter elements that are absent in the slob57 promoter coordinately drive robust expression of a luciferase vector by the slob71 promoter in vitro. In addition, we visualized the expression patterns of the slob57 and slob71 promoters in vivo and found clear spatiotemporal differences in promoter activity. SLOB57 is expressed prominently in adult PI neurons, whereas larval motor neurons exclusively express SLOB71. In contrast, at the larval neuromuscular junction, SLOB57 expression appears to be restricted mainly to a subset of glial cells. Our results illustrate how the use of alternative transcriptional start sites within an ion channel modulator locus coupled with functionally relevant alternative splicing can be used to fine-tune neuronal excitability in a cell-specific manner.


Subject(s)
Brain/metabolism , Drosophila Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Protein Isoforms/metabolism , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Larva/genetics , Larva/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Potassium Channels/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Synaptic Transmission/physiology
6.
PLoS One ; 6(8): e23343, 2011.
Article in English | MEDLINE | ID: mdl-21850269

ABSTRACT

There is ample evidence that ion channel modulation by accessory proteins within a macromolecular complex can regulate channel activity and thereby impact neuronal excitability. However, the downstream consequences of ion channel modulation remain largely undetermined. The Drosophila melanogaster large conductance calcium-activated potassium channel SLOWPOKE (SLO) undergoes modulation via its binding partner SLO-binding protein (SLOB). Regulation of SLO by SLOB influences the voltage dependence of SLO activation and modulates synaptic transmission. SLO and SLOB are expressed especially prominently in median neurosecretory cells (mNSCs) in the pars intercerebralis (PI) region of the brain; these cells also express and secrete Drosophila insulin like peptides (dILPs). Previously, we found that flies lacking SLOB exhibit increased resistance to starvation, and we reasoned that SLOB may regulate aspects of insulin signaling and metabolism. Here we investigate the role of SLOB in metabolism and find that slob null flies exhibit changes in energy storage and insulin pathway signaling. In addition, slob null flies have decreased levels of dilp3 and increased levels of takeout, a gene known to be involved in feeding and metabolism. Targeted expression of SLOB to mNSCs rescues these alterations in gene expression, as well as the metabolic phenotypes. Analysis of fly lines mutant for both slob and slo indicate that the effect of SLOB on metabolism and gene expression is via SLO. We propose that modulation of SLO by SLOB regulates neurotransmission in mNSCs, influencing downstream insulin pathway signaling and metabolism.


Subject(s)
Drosophila Proteins/metabolism , Insulin/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium Channels/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Brain/metabolism , Drosophila , Drosophila Proteins/genetics , Glucose/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Potassium Channels/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Trehalose/metabolism
7.
J Gen Physiol ; 137(2): 225-38, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21282401

ABSTRACT

Modulation of ion channels by regulatory proteins within the same macromolecular complex is a well-accepted concept, but the physiological consequences of such modulation are not fully understood. Slowpoke (Slo), a potassium channel critical for action potential repolarization and transmitter release, is regulated by Slo channel-binding protein (Slob), a Drosophila melanogaster Slo (dSlo) binding partner. Slob modulates the voltage dependence of dSlo channel activation in vitro and exerts similar effects on the dSlo channel in Drosophila central nervous system neurons in vivo. In addition, Slob modulates action potential duration in these neurons. Here, we investigate further the functional consequences of the modulation of the dSlo channel by Slob in vivo, by examining larval neuromuscular synaptic transmission in flies in which Slob levels have been altered. In Slob-null flies generated through P-element mutagenesis, as well as in Slob knockdown flies generated by RNA interference (RNAi), we find an enhancement of synaptic transmission but no change in the properties of the postsynaptic muscle cell. Using targeted transgenic rescue and targeted expression of Slob-RNAi, we find that Slob expression in neurons (but not in the postsynaptic muscle cell) is critical for its effects on synaptic transmission. Furthermore, inhibition of dSlo channel activity abolishes these effects of Slob. These results suggest that presynaptic Slob, by regulating dSlo channel function, participates in the modulation of synaptic transmission.


Subject(s)
Drosophila Proteins/metabolism , Potassium Channels/metabolism , Synaptic Transmission/physiology , Animals , Animals, Genetically Modified , Carrier Proteins , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Neurons/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , RNA Interference
8.
Neuron ; 65(5): 670-81, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20223202

ABSTRACT

An understanding of sleep requires the identification of distinct cellular circuits that mediate the action of specific sleep:wake-regulating molecules, but such analysis has been very limited. We identify here a circuit that underlies the wake-promoting effects of octopamine in Drosophila. Using MARCM, we identified the ASM cells in the medial protocerebrum as the wake-promoting octopaminergic cells. We then blocked octopamine signaling in random areas of the fly brain and mapped the postsynaptic effect to insulin-secreting neurons of the pars intercerebralis (PI). These PI neurons show altered potassium channel function as well as an increase in cAMP in response to octopamine, and genetic manipulation of their electrical excitability alters sleep:wake behavior. Effects of octopamine on sleep:wake are mediated by the cAMP-dependent isoform of the OAMB receptor. These studies define the cellular and molecular basis of octopamine action and suggest that the PI is a sleep:wake-regulating neuroendocrine structure like the mammalian hypothalamus.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Nerve Net/drug effects , Octopamine/pharmacology , Sleep/drug effects , Wakefulness/drug effects , Animals , Animals, Genetically Modified , Behavior, Animal , Circadian Rhythm/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Electric Stimulation/methods , Green Fluorescent Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Nerve Net/physiology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Sleep/physiology , Tetraethylammonium/pharmacology , Wakefulness/physiology
9.
J Neurosci ; 29(42): 13328-37, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19846720

ABSTRACT

Molecular details of ion channel interactions with modulatory subunits have been investigated widely in transfected cells, but the physiological roles of ion channel modulatory protein complexes in native neurons remain largely unexplored. The Drosophila large-conductance calcium-activated potassium channel (dSlo) binds to and is modulated by its binding partner Slob. We have constructed flies in which Slob expression is manipulated by P-element mutagenesis, or by transgenic expression of Slob protein or Slob-RNAi. In vivo recordings of both macroscopic and single dSlo channel currents in identified neurosecretory neurons in the pars intercerebralis (PI) region of the Drosophila brain reveal that whole-cell potassium current and properties of single dSlo channels are modulated by Slob expression level. Furthermore, Slob genotype influences action potential duration in vivo. This unprecedented combination of current-clamp, macroscopic-current, and single-channel recordings from neurons in brains of living flies defines a critical role for an ion channel modulatory protein complex in the control of neuronal excitability. We show further that Slob-null flies exhibit significantly longer lifespan than controls under conditions of complete food deprivation. Crosses with deficiency lines demonstrate that this enhanced resistance to starvation-induced death maps close to the slob locus. Together, these results indicate that Slob may serve a novel regulatory function in feeding behavior, possibly by influencing the excitability of the PI neurons.


Subject(s)
Drosophila Proteins/physiology , Feeding Behavior/physiology , Gene Expression Regulation/genetics , Neurons/physiology , Potassium Channels/physiology , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Biophysical Phenomena/drug effects , Biophysical Phenomena/physiology , Cerebellum/cytology , Cerebellum/metabolism , Drosophila , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Electric Conductivity , Electric Stimulation/methods , Food Deprivation/physiology , Green Fluorescent Proteins/genetics , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutagenesis , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , Potassium Channels/deficiency , Potassium Channels/genetics , RNA, Small Interfering/genetics , Starvation/genetics , Tetraethylammonium/pharmacology
10.
Neuron ; 59(2): 188-9, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18667146

ABSTRACT

Ion channels are modulated by multiple molecular mechanisms. In this issue of Neuron, Pietrzykowski et al. expand the mechanistic repertoire by demonstrating that ethanol-induced microRNA can modulate the pattern of mRNA splice variants from which BK potassium channels are constructed. Because BK channels are important targets of ethanol, this finding has implications for mechanisms of ethanol sensitivity and tolerance.


Subject(s)
Ion Channel Gating/physiology , Ion Channels/physiology , MicroRNAs/physiology , Animals , Humans , RNA, Messenger/physiology , Second Messenger Systems/physiology
11.
J Neurophysiol ; 99(5): 2736-40, 2008 May.
Article in English | MEDLINE | ID: mdl-18385479

ABSTRACT

Voltage-gated KCNQ potassium channels are responsible for slowly activating potassium currents in heart, brain, and other tissues. Functional defects of KCNQ channels are linked with many diseases, including epilepsy and cardiac arrhythmias. Therefore KCNQ potassium channels have been widely studied, especially in the CNS. We have identified Drosophila CG11963, which encodes a protein orthologous to the beta subunit of mammalian succinyl-CoA synthetase (SCS, also known as succinate thiokinase), as a novel modulator of Drosophila KCNQ channels. Direct interaction of CG11963 and dKCNQ was demonstrated by yeast two-hybrid screen and coimmunoprecipitation. Cell surface biotinylation experiments further confirmed that CG11963 resides on the plasma membrane of tsA-201 cells. Coexpression of CG11963 with dKCNQ shifts the conductance-voltage (G-V) relationship of dKCNQ channels to more positive membrane potentials in Chinese hamster ovary (CHO) cells. Moreover, directly dialyzing glutathione S-transferase fusion CG11963 protein into CHO cells also shifts the dKCNQ G-V curve rightward. The effect of CG11963 persists in the presence of 1 mM adenosine triphosphate (ATP), a substrate of SCS. Taken together, our data define CG11963 as a new dKCNQ-binding protein capable of modulating the properties of the channel. Our evidence suggests that this modulation is mediated by direct interaction of CG11963 with the channel and is not dependent on ATP.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , KCNQ Potassium Channels/physiology , Succinate-CoA Ligases/physiology , Adenosine Triphosphate/physiology , Amino Acid Sequence , Animals , Biotinylation , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiology , Glutathione Transferase/metabolism , Molecular Sequence Data , Patch-Clamp Techniques
12.
J Gen Physiol ; 128(5): 583-91, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17074977

ABSTRACT

Slob57 is an ion channel auxiliary protein that binds to and modulates the Drosophila Slowpoke calcium-dependent potassium channel (dSlo). We reported recently that residues 1-39 of Slob57 comprise the key domain that both causes dSlo inactivation and shifts its voltage dependence of activation to more depolarized voltages. In the present study we show that removal of residues 2-6 from Slob57 abolishes the inactivation, but the ability of Slob57 to rightward shift the voltage dependence of activation of dSlo remains. A synthetic peptide corresponding in sequence to residues 1-6 of Slob57 blocks dSlo in a voltage- and dose-dependent manner. Two Phe residues and at least one Lys residue in this peptide are required for the blocking action. These data indicate that the amino terminus of Slob57 directly blocks dSlo, thereby leading to channel inactivation. Further truncation to residue Arg(16) eliminates the modulation of voltage dependence of activation. Thus these two modulatory actions of Slob57 are independent. Mutation within the calcium bowl of dSlo greatly reduces its calcium sensitivity (Bian, S., I. Favre, and E. Moczydlowski. 2001. Proc. Natl. Acad. Sci. USA. 98:4776-4781). We found that Slob57 still causes inactivation of this mutant channel, but does not shift its voltage dependence of activation. This result confirms further the independence of the inactivation and the voltage shift produced by Slob57. It also suggests that the voltage shift requires high affinity Ca(2+) binding to an intact calcium bowl. Furthermore, Slob57 inhibits the shift in the voltage dependence of activation of dSlo evoked by Ca(2+), and this inhibition by Slob57 is greater at higher free Ca(2+) concentrations. These results implicate distinct calcium-dependent and -independent mechanisms in the modulation of dSlo by Slob.


Subject(s)
Drosophila Proteins/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Potassium Channels/physiology , Amino Acids/analysis , Amino Acids/genetics , Animals , CHO Cells , Calcium/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Drosophila , Drosophila Proteins/chemistry , Drosophila Proteins/drug effects , Drosophila Proteins/genetics , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/genetics , Membrane Potentials/physiology , Mutation/genetics , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channels/chemistry , Potassium Channels/drug effects , Potassium Channels/genetics
13.
Nat Neurosci ; 9(3): 305-10, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16498425

ABSTRACT

The pore-forming subunits of many ion channels exist in the membrane as one component of a regulatory protein complex, which may also contain one or more signaling proteins that contribute to the modulation of channel properties. Here I review this field, with emphasis on several different kinds of neuronal potassium channels for which the evidence for ion channel signaling complexes is most compelling. A key challenge for the future is to determine the roles of such signaling protein complexes in neuronal physiology and behavior.


Subject(s)
Ion Channels/metabolism , Membrane Proteins/metabolism , Nervous System/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Calcium Signaling/physiology , Humans , Macromolecular Substances , Potassium Channels/metabolism , Protein Kinases/metabolism
14.
J Neurophysiol ; 95(3): 1957-65, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339006

ABSTRACT

Slob binds to and modulates the Drosophila Slowpoke (dSlo) calcium-activated potassium channel and also recruits the ubiquitous signaling protein 14-3-3 to the channel regulatory complex. RT-PCR reveals the presence of multiple slob transcripts in Drosophila heads. The transcripts are predicted to encode proteins that we call Slob51 (kDa), Slob57, Slob65, and Slob71. Slob51 and Slob65 are splice variants that lack a motif important for the binding of 14-3-3. Previous microarray analyses demonstrated the circadian cycling of slob mRNA, and we show by quantitative PCR that more than one transcript cycles in fly heads. Using in situ hybridization, we observe differences in the expression patterns of the different transcripts. Immunohistochemistry on Drosophila heads reveals Slob71/65 protein to be enriched in the lateral neurons, in contrast to Slob57/51 protein, which is expressed most prominently in the pars intercerebralis neurons and dorsal giant interneurons. Using a heterologous expression system, we show that different Slobs bind to different extents to dSlo and 14-3-3. These data reveal an unexpected diversity of the dSlo/Slob/14-3-3 dynamic regulatory complex.


Subject(s)
14-3-3 Proteins/metabolism , Brain/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Animals , Drosophila Proteins/classification , Gene Expression Profiling , Large-Conductance Calcium-Activated Potassium Channels/classification , Potassium Channels/classification , Protein Interaction Mapping
15.
Proc Natl Acad Sci U S A ; 102(45): 16454-9, 2005 Nov 08.
Article in English | MEDLINE | ID: mdl-16263935

ABSTRACT

KCNQ2 and KCNQ3 ion channel pore-forming subunits coassemble to form a heteromeric voltage-gated potassium channel that underlies the neuronal M-current. We and others showed that calmodulin (CaM) binds to specific sequence motifs in the C-terminal domain of KCNQ2 and KCNQ3. We also found that a fusion protein containing a KCNQ2 CaM-binding motif, coexpressed with KCNQ2 and KCNQ3, competes with the full-length KCNQ2 channel for CaM binding and thereby decreases KCNQ2/3 current density in heterologous cells. We have explored the importance of CaM binding for the generation of the native M-current and regulation of membrane excitability in rat hippocampal neurons in primary cell culture. M-current properties were studied in cultured neurons by using whole-cell patch clamp recording. The M-current density is lower in neurons expressing the CaM-binding motif fusion protein, as compared to control neurons transfected with vector alone. In contrast, no change in M-current density is observed in cells transfected with a mutant fusion protein that is unable to bind CaM. The CaM-binding fusion protein does not influence the rapidly inactivating A-current or the large conductance calcium-activated potassium channel-mediated fast spike afterhyperpolarization in neurons in which the M-current is suppressed. Furthermore, the CaM-binding fusion protein, but not the nonbinding mutant, increases both the number of action potentials evoked by membrane depolarization and the size of the spike afterdepolarization. These results suggest that CaM binding regulates M-channel function and membrane excitability in the native neuronal environment.


Subject(s)
Calmodulin/physiology , KCNQ2 Potassium Channel/physiology , Neurons/physiology , Peptide Fragments/physiology , Recombinant Fusion Proteins/physiology , Animals , Cells, Cultured , Female , KCNQ3 Potassium Channel/physiology , Membrane Potentials , Pregnancy , Rats , Rats, Sprague-Dawley
16.
J Neurosci ; 25(44): 10147-56, 2005 Nov 02.
Article in English | MEDLINE | ID: mdl-16267222

ABSTRACT

The mammalian voltage-dependent KCNQ channels are responsible for distinct types of native potassium currents and are associated with several human diseases. We cloned a novel Drosophila KCNQ channel (dKCNQ) based on its sequence homology to the mammalian genes. When expressed in Chinese hamster ovary cells, dKCNQ gives rise to a slowly activating and slowly deactivating current that activates in the subthreshold voltage range. Like the M-current produced by mammalian KCNQ channels, dKCNQ current is sensitive to the KCNQ-specific blocker linopirdine and is suppressed by activation of a muscarinic receptor. dKCNQ is also similar to the mammalian channels in that it binds calmodulin (CaM), and CaM binding is necessary to produce functional currents. In situ hybridization analysis demonstrates that dKCNQ mRNA is present in brain cortical neurons, the cardia (proventriculus), and the nurse cells and oocytes of the ovary. We generated mutant flies with deletions in the genomic sequence of dKCNQ. Embryos produced by homozygous deletion females exhibit disorganized nuclei and fail to hatch, suggesting strongly that a maternal contribution of dKCNQ protein and/or mRNA is essential for early embryonic development.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , KCNQ Potassium Channels/physiology , Animals , CHO Cells , Cricetinae , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/genetics , Male , Mice , Potassium Channel Blockers/pharmacology , RNA, Messenger/physiology
17.
J Neurosci ; 25(35): 7934-43, 2005 Aug 31.
Article in English | MEDLINE | ID: mdl-16135750

ABSTRACT

We have cloned and characterized mouse and human variants of MONaKA, a novel protein that interacts with and modulates the plasma membrane Na,K-ATPase. MONaKA was cloned based on its sequence homology to the Drosophila Slowpoke channel-binding protein dSlob, but mouse and human MONaKA do not bind to mammalian Slowpoke channels. At least two splice variants of MONaKA exist; the splicing is conserved perfectly between mouse and human, suggesting that it serves some important function. Both splice variants of MONaKA are expressed widely throughout the CNS and peripheral nervous system, with different splice variant expression ratios in neurons and glia. A yeast two-hybrid screen with MONaKA as bait revealed that it binds tightly to the beta1 and beta3 subunits of the Na,K-ATPase. The association between MONaKA and Na,K-ATPase beta subunits was confirmed further by coimmunoprecipitation from transfected cells, mouse brain, and cultured mouse astrocytes. A glutathione S-transferase-MONaKA fusion protein inhibits Na,K-ATPase activity from whole brain or cultured astrocytes. Furthermore, transfection of MONaKA inhibits 86Rb+ uptake via the Na,K-ATPase in intact cells. These results are consistent with the hypothesis that MONaKA modulates brain Na,K-ATPase and may thereby participate in the regulation of electrical excitability and synaptic transmission.


Subject(s)
Cell Membrane/enzymology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Alternative Splicing , Animals , Cell Membrane/genetics , Cells, Cultured , Cloning, Molecular/methods , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Pathways/enzymology , Neural Pathways/physiology , Protein Serine-Threonine Kinases , Rats , Sodium-Potassium-Exchanging ATPase/genetics , Synapses/enzymology , Synapses/physiology
18.
J Gen Physiol ; 125(6): 631-40, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15897294

ABSTRACT

The Drosophila Slowpoke calcium-dependent potassium channel (dSlo) binding protein Slob was discovered by a yeast two-hybrid screen using the carboxy-terminal tail region of dSlo as bait. Slob binds to and modulates the dSlo channel. We have found that there are several Slob proteins, resulting from multiple translational start sites and alternative splicing, and have named them based on their molecular weights (in kD). The larger variants, which are initiated at the first translational start site and are called Slob71 and Slob65, shift the voltage dependence of dSlo activation, measured by the whole cell conductance-voltage relationship, to the left (less depolarized voltages). Slob53 and Slob47, initiated at the third translational start site, also shift the dSlo voltage dependence to the left. In contrast, Slob57 and Slob51, initiated at the second translational start site, shift the conductance-voltage relationship of dSlo substantially to more depolarized voltages, cause an apparent dSlo channel inactivation, and increase the deactivation rate of the channel. These results indicate that the amino-terminal region of Slob plays a critical role in its modulation of dSlo.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Potassium Channels, Calcium-Activated/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Amino Acid Sequence/physiology , Animals , CHO Cells , Cloning, Molecular , Cricetinae , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Drosophila , Drosophila Proteins/antagonists & inhibitors , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels , Molecular Weight
19.
J Neurophysiol ; 93(3): 1393-405, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15496493

ABSTRACT

From its position in presynaptic nerve terminals, the large conductance Ca(2+)-activated K+ channel, Slo, regulates neurotransmitter release. Several other ion channels known to control neurotransmitter release have been implicated in physical interactions with the neurotransmitter release machinery. For example, the Ca(v)2.2 (N-type) Ca2+ channel binds to and is modulated by syntaxin-1A and SNAP-25. Furthermore, a close juxtaposition of Slo and Ca(v)2.2 is presumed to be necessary for functional coupling between the two channels, which has been shown in neurons. We report that Slo exhibits a strong association with syntaxin-1A. Robust co-immunoprecipitation of Slo and syntaxin-1A occurs from transfected HEK293 cells as well as from brain. However, despite this strong interaction and the known association between syntaxin-1A and the II-III loop of Ca(v)2.2, these three proteins do not co-immunoprecipitate in a trimeric complex from transfected HEK293 cells. The Slo-syntaxin-1A co-immunoprecipitation is not significantly influenced by [Ca2+]. Multiple relatively weak interactions may sum up to a tight physical coupling of full-length Slo with syntaxin-1A: the C-terminal tail and the S0-S1 loop of Slo each co-immunoprecipitate with syntaxin-1A. The presence of syntaxin-1A leads to reduced Slo channel activity due to an increased V(1/2) for activation in 100 nM, 1 muM, and 10 microM Ca2+, reduced voltage-sensitivity in 1 microM Ca2+, and slower rates of activation in 10 microM Ca2+. Potential physiological consequences of the interaction between Slo and syntaxin-1A include enhanced excitability through modulation of Slo channel activity and reduced neurotransmitter release due to disruption of syntaxin-1A binding to the Ca(v)2.2 II-III loop.


Subject(s)
Antigens, Surface/metabolism , Calcium Channels/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Calcium-Activated/metabolism , Blotting, Western/methods , Brain/metabolism , Calcium/metabolism , Calcium/pharmacology , Cell Line , Cloning, Molecular/methods , Dose-Response Relationship, Drug , Drug Interactions , Humans , Immunoprecipitation/methods , Large-Conductance Calcium-Activated Potassium Channels , Macromolecular Substances , Membrane Potentials/drug effects , Patch-Clamp Techniques/methods , Protein Binding , Protein Structure, Tertiary/physiology , Qa-SNARE Proteins , Syntaxin 1 , Transfection/methods
20.
Neurosci Lett ; 365(1): 33-8, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15234468

ABSTRACT

The slowpoke channel binding protein Slob from Drosophila melanogaster contains a putative protein kinase domain within its amino acid sequence. We find that Slob exhibits weak and barely detectable protein kinase activity in vitro, as evidenced by autophosphorylation and by phosphorylation of exogenously added histone as substrate. The phosphorylation of histone is enhanced markedly when Slob is pretreated with the catalytic subunit of cyclic AMP-dependent protein kinase (PKAc). Mass spectrometric and mutational analysis demonstrates that the major site of phosphorylation by PKAc within Slob is serine 54. The enhancement of Slob kinase activity by PKAc pretreatment is eliminated when serine 54 in Slob is mutated to alanine (S54A). Furthermore, Slob kinase activity is enhanced in an S54E mutant that mimics phosphorylation at serine 54, and there is no further enhancement of S54E Slob kinase activity by pretreatment with PKAc. The results are consistent with the hypothesis that Slob exhibits regulatable protein kinase activity, whose activity is enhanced by phosphorylation at serine 54.


Subject(s)
Drosophila Proteins/metabolism , Potassium Channels/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Electrophoresis, Polyacrylamide Gel , Histones/metabolism , Mass Spectrometry , Phosphorylation , Point Mutation , Potassium Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...