Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-30846936

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play a critical role in synaptic signaling, and alterations in the synaptic/extrasynaptic NMDAR balance affect neuronal survival. Studies have shown enhanced extrasynaptic GluN2B-type NMDAR (2B-NMDAR) activity in striatal neurons in the YAC128 mouse model of Huntington disease (HD), resulting in increased cell death pathway activation contributing to striatal vulnerability to degeneration. However, the mechanism(s) of altered GluN2B trafficking remains unclear. Previous work shows that GluN2B palmitoylation on two C-terminal cysteine clusters regulates 2B-NMDAR trafficking to the surface membrane and synapses in cortical neurons. Notably, two palmitoyl acyltransferases (PATs), zDHHC17 and zDHHC13, also called huntingtin-interacting protein 14 (HIP14) and HIP14-like (HIP14L), directly interact with the huntingtin protein (Htt), and mutant Htt disrupts this interaction. Here, we investigated whether GluN2B palmitoylation is involved in enhanced extrasynaptic surface expression of 2B-NMDARs in YAC128 striatal neurons and whether this process is regulated by HIP14 or HIP14L. We found reduced GluN2B palmitoylation in YAC128 striatum, specifically on cysteine cluster II. Consistent with that finding, the palmitoylation-deficient GluN2B Cysteine cluster II mutant exhibited enhanced, extrasynaptic surface expression in striatal neurons from wild-type mice, mimicking increased extrasynaptic 2B-NMDAR observed in YAC128 cultures. We also found that HIP14L palmitoylated GluN2B cysteine cluster II. Moreover, GluN2B palmitoylation levels were reduced in striatal tissue from HIP14L-deficient mice, and siRNA-mediated HIP14L knockdown in cultured neurons enhanced striatal neuronal GluN2B surface expression and susceptibility to NMDA toxicity. Thus, altered regulation of GluN2B palmitoylation levels by the huntingtin-associated PAT HIP14L may contribute to the cell death-signaling pathways underlying HD.

2.
Nat Commun ; 7: 11251, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052848

ABSTRACT

It has become well accepted that Huntington disease (HD) is associated with impaired glutamate uptake, resulting in a prolonged time-course of extracellular glutamate that contributes to excitotoxicity. However, the data supporting this view come largely from work in synaptosomes, which may overrepresent nerve-terminal uptake over astrocytic uptake. Here, we quantify real-time glutamate dynamics in HD mouse models by high-speed imaging of an intensity-based glutamate-sensing fluorescent reporter (iGluSnFR) and electrophysiological recordings of synaptically activated transporter currents in astrocytes. These techniques reveal a disconnect between the results obtained in synaptosomes and those in situ. Exogenous glutamate uptake is impaired in synaptosomes, whereas real-time measures of glutamate clearance in the HD striatum are normal or even accelerated, particularly in the aggressive R6/2 model. Our results highlight the importance of quantifying glutamate dynamics under endogenous release conditions, and suggest that the widely cited uptake impairment in HD does not contribute to pathogenesis.


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Huntington Disease/metabolism , Optical Imaging/methods , Synapses/metabolism , Animals , Astrocytes/pathology , Biological Transport , Corpus Striatum/pathology , Dependovirus/genetics , Disease Models, Animal , Genes, Reporter , Genetic Vectors , Humans , Huntington Disease/pathology , Male , Membrane Potentials/physiology , Mice , Mice, Transgenic , Synaptosomes/metabolism
3.
Neuron ; 86(3): 680-95, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25913858

ABSTRACT

22q11.2 deletion carriers show specific cognitive deficits, and ∼30% of them develop schizophrenia. One of the disrupted genes is ZDHHC8, which encodes for a palmitoyltransferase. We show that Zdhhc8-deficient mice have reduced palmitoylation of proteins that regulate axonal growth and branching. Analysis of axonal projections of pyramidal neurons from both Zdhhc8-deficient and Df(16)A(+/-) mice, which model the 22q11.2 deletion, revealed deficits in axonal growth and terminal arborization, which can be prevented by reintroduction of active ZDHHC8 protein. Impaired terminal arborization is accompanied by a reduction in the strength of synaptic connections and altered functional connectivity and working memory. The effect of ZDHHC8 is mediated in part via Cdc42-dependent modulation of Akt/Gsk3ß signaling at the tip of the axon and can be reversed by pharmacologically decreasing Gsk3ß activity during postnatal brain development. Our findings provide valuable mechanistic insights into the cognitive and psychiatric symptoms associated with a schizophrenia-predisposing mutation.


Subject(s)
Alzheimer Disease/pathology , Axons/pathology , Brain/pathology , DiGeorge Syndrome/pathology , Acyltransferases/deficiency , Acyltransferases/genetics , Age Factors , Alzheimer Disease/genetics , Animals , Animals, Newborn , Brain/embryology , Brain/metabolism , Channelrhodopsins , DiGeorge Syndrome/genetics , Disease Models, Animal , Embryo, Mammalian , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurons/pathology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Signal Transduction/genetics , Synapsins/metabolism
4.
J Biol Chem ; 289(6): 3518-28, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24347167

ABSTRACT

Huntington disease is associated with early alterations in corticostriatal synaptic function that precede cell death, and it is postulated that ameliorating such changes may delay clinical onset and/or prevent neurodegeneration. Although many of these synaptic alterations are thought to be attributable to a toxic gain of function of the mutant huntingtin protein, the role that nonpathogenic huntingtin (HTT) plays in synaptic function is relatively unexplored. Here, we compare the immunocytochemical localization of a major postsynaptic scaffolding protein, PSD-95, in striatal neurons from WT mice and mice overexpressing HTT with 18 glutamine repeats (YAC18, nonpathogenic). We found that HTT overexpression resulted in a palmitoylation- and BDNF-dependent increase in PSD-95 clustering at synaptic sites in striatal spiny projection neurons (SPNs) co-cultured with cortical neurons. Surprisingly, the latter effect was mediated presynaptically, as HTT overexpression in cortical neurons alone was sufficient to increase PSD-95 clustering in the postsynaptic SPNs. In contrast, antisense oligonucleotide knockdown of HTT in WT co-cultures resulted in a significant reduction of PSD-95 clustering in SPNs. Notably, despite these bidirectional changes in PSD-95 clustering, we did not observe an alteration in basal electrophysiological measures of AMPA and NMDA receptors. Thus, unlike in previous studies in the hippocampus, enhanced or decreased PSD-95 clustering alone was insufficient to drive AMPA or NMDA receptors into or out of SPN synapses. In all, our results demonstrate that nonpathogenic HTT can indeed influence synaptic protein localization and uncover a novel role of HTT in PSD-95 distribution.


Subject(s)
Corpus Striatum/metabolism , Guanylate Kinases/metabolism , Lipoylation/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Post-Synaptic Density/metabolism , Animals , Corpus Striatum/cytology , Disks Large Homolog 4 Protein , Gene Knockdown Techniques , Guanylate Kinases/genetics , Hippocampus/cytology , Hippocampus/metabolism , Huntingtin Protein , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Nuclear Proteins/genetics , Post-Synaptic Density/genetics , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
5.
J Neurosci ; 32(21): 7119-36, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623657

ABSTRACT

NMDA receptor-dependent long-term potentiation (LTP) and depression (LTD) are forms of synaptic plasticity underlying learning and memory that are expressed through increases and decreases, respectively, in dendritic spine size and AMPA receptor (AMPAR) phosphorylation and postsynaptic localization. The A-kinase anchoring protein 79/150 (AKAP79/150) signaling scaffold regulates AMPAR phosphorylation, channel activity, and endosomal trafficking associated with LTP and LTD. AKAP79/150 is targeted to dendritic spine plasma membranes by an N-terminal polybasic domain that binds phosphoinositide lipids, F-actin, and cadherin cell adhesion molecules. However, we do not understand how regulation of AKAP targeting controls AMPAR endosomal trafficking. Here, we report that palmitoylation of the AKAP N-terminal polybasic domain targets it to postsynaptic lipid rafts and dendritic recycling endosomes. AKAP palmitoylation was regulated by seizure activity in vivo and LTP/LTD plasticity-inducing stimuli in cultured rat hippocampal neurons. With chemical LTP induction, we observed AKAP79 dendritic spine recruitment that required palmityolation and Rab11-regulated endosome recycling coincident with spine enlargement and AMPAR surface delivery. Importantly, a palmitoylation-deficient AKAP79 mutant impaired regulation of spine size, endosome recycling, AMPAR trafficking, and synaptic potentiation. These findings emphasize the emerging importance of palmitoylation in controlling synaptic function and reveal novel roles for the AKAP79/150 signaling complex in dendritic endosomes.


Subject(s)
A Kinase Anchor Proteins/physiology , Dendrites/metabolism , Endosomes/metabolism , Neuronal Plasticity/physiology , Protein Transport/physiology , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Dendritic Spines/ultrastructure , Female , Gene Knockdown Techniques , Hippocampus/metabolism , Hippocampus/physiology , Kainic Acid/pharmacology , Lipoylation/physiology , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, AMPA/metabolism , Seizures/chemically induced , Seizures/metabolism , Seizures/physiopathology
6.
Hum Mol Genet ; 20(17): 3356-65, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21636527

ABSTRACT

Huntington disease (HD) is caused by polyglutamine expansion in the huntingtin (HTT) protein. Huntingtin-interacting protein 14 (HIP14), one of 23 DHHC domain-containing palmitoyl acyl transferases (PATs), binds to HTT and robustly palmitoylates HTT at cysteine 214. Mutant HTT exhibits reduced palmitoylation and interaction with HIP14, contributing to the neuronal dysfunction associated with HD. In this study, we confirmed that, among 23 DHHC PATs, HIP14 and its homolog DHHC-13 (HIP14L) are the two major PATs that palmitoylate HTT. Wild-type HTT, in addition to serving as a palmitoylation substrate, also modulates the palmitoylation of HIP14 itself. In vivo, HIP14 palmitoylation is decreased in the brains of mice lacking one HTT allele (hdh+/-) and is further reduced in mouse cortical neurons treated with HTT antisense oligos (HTT-ASO) that knockdown HTT expression by ∼95%. Previously, it has been shown that palmitoylation of DHHC proteins may affect their enzymatic activity. Indeed, palmitoylation of SNAP25 by HIP14 is potentiated in vitro in the presence of wild-type HTT. This influence of HTT on HIP14 activity is lost in the presence of CAG expansion. Furthermore, in both brains of hdh+/- mice and neurons treated with HTT-ASO, we observe a significant reduction in palmitoylation of endogenous SNAP25 and GluR1, synaptic proteins that are substrates of HIP14, suggesting wild-type HTT also influences HIP14 enzymatic activity in vivo. This study describes an important biochemical function for wild-type HTT modulation of HIP14 palmitoylation and its enzymatic activity.


Subject(s)
Acyltransferases/metabolism , Huntington Disease/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Acyltransferases/genetics , Animals , Blotting, Western , Cells, Cultured , Huntingtin Protein , Huntington Disease/genetics , Lipoylation , Mice , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Protein Binding , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Two-Hybrid System Techniques
7.
Neurobiol Dis ; 40(1): 207-15, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685337

ABSTRACT

Excitotoxicity plays a key role in the selective vulnerability of striatal neurons in Huntington disease (HD). Decreased glutamate uptake by glial cells could account for the excess glutamate at the synapse in patients as well as animal models of HD. The major molecule responsible for clearing glutamate at the synapses is glial glutamate transporter GLT-1. In this study, we show that GLT-1 is palmitoylated at cysteine38 (C38) and further, that this palmitoylation is drastically reduced in HD models both in vitro and in vivo. Palmitoylation is required for normal GLT-1 function. Blocking palmitoylation either with the general palmitoylation inhibitor, 2-bromopalmitate, or with a GLT-1 C38S mutation, severely impairs glutamate uptake activity. In addition, GLT-1-mediated glutamate uptake is indeed impaired in the YAC128 HD mouse brain, with the defect in the striatum evident as early as 3 months prior to obvious neuropathological findings, and in both striatum and cortex at 12 months. These phenotypes are not a result of changes in GLT1 protein expression, suggesting a crucial role of palmitoylation in GLT-1 function. Thus, it appears that impaired GLT-1 palmitoylation is present early in the pathogenesis of HD, and may influence decreased glutamate uptake, excitotoxicity, and ultimately, neuronal cell death in HD.


Subject(s)
Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/metabolism , Huntington Disease/metabolism , Lipoylation/physiology , Neuroglia/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cysteine/genetics , Cysteine/metabolism , Disease Models, Animal , Down-Regulation/physiology , Excitatory Amino Acid Transporter 2/physiology , Glutamic Acid/metabolism , Huntington Disease/etiology , Huntington Disease/genetics , Lipoylation/drug effects , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Rats
8.
J Biol Chem ; 285(7): 4621-8, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-19955568

ABSTRACT

The Golgi-specific zinc finger protein GODZ (palmitoyl acyltransferase/DHHC-3) mediates the palmitoylation and post-translational modification of many protein substrates that regulate membrane-protein interactions. Here, we show that GODZ also mediates Ca(2+) transport in expressing Xenopus laevis oocytes. Two-electrode voltage-clamp, fluorescence, and (45)Ca(2+) isotopic uptake determinations demonstrated voltage- and concentration-dependent, saturable, and substrate-inhibitable Ca(2+) transport in oocytes expressing GODZ cRNA but not in oocytes injected with water alone. Moreover, we show that GODZ-mediated Ca(2+) transport is regulated by palmitoylation, as the palmitoyl acyltransferase inhibitor 2-bromopalmitate or alteration of the acyltransferase DHHC motif (GODZ-DHHS) diminished GODZ-mediated Ca(2+) transport by approximately 80%. The GODZ mutation V61R abolished Ca(2+) transport but did not affect palmitoyl acyltransferase activity. Coexpression of GODZ-V61R with GODZ-DHHS restored GODZ-DHHS-mediated Ca(2+) uptake to values observed with wild-type GODZ, excluding an endogenous effect of palmitoylation. Coexpression of an independent palmitoyl acyltransferase (HIP14) with the GODZ-DHHS mutant also rescued Ca(2+) transport. HIP14 did not mediate Ca(2+) transport when expressed alone. Immunocytochemistry studies showed that GODZ and HIP14 co-localized to the Golgi and the same post-Golgi vesicles, suggesting that heteropalmitoylation might play a physiological role in addition to a biochemical function. We conclude that GODZ encodes a Ca(2+) transport protein in addition to its ability to palmitoylate protein substrates.


Subject(s)
Acyltransferases/metabolism , Calcium/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Acyltransferases/chemistry , Acyltransferases/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Microscopy, Confocal , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oocytes , Protein Transport/genetics , Protein Transport/physiology , Xenopus/genetics , Xenopus/metabolism
9.
Nature ; 456(7224): 904-9, 2008 Dec 18.
Article in English | MEDLINE | ID: mdl-19092927

ABSTRACT

Palmitoylation regulates diverse aspects of neuronal protein trafficking and function. Here a global characterization of rat neural palmitoyl-proteomes identifies most of the known neural palmitoyl proteins-68 in total, plus more than 200 new palmitoyl-protein candidates, with further testing confirming palmitoylation for 21 of these candidates. The new palmitoyl proteins include neurotransmitter receptors, transporters, adhesion molecules, scaffolding proteins, as well as SNAREs and other vesicular trafficking proteins. Of particular interest is the finding of palmitoylation for a brain-specific Cdc42 splice variant. The palmitoylated Cdc42 isoform (Cdc42-palm) differs from the canonical, prenylated form (Cdc42-prenyl), both with regard to localization and function: Cdc42-palm concentrates in dendritic spines and has a special role in inducing these post-synaptic structures. Furthermore, assessing palmitoylation dynamics in drug-induced activity models identifies rapidly induced changes for Cdc42 as well as for other synaptic palmitoyl proteins, suggesting that palmitoylation may participate broadly in the activity-driven changes that shape synapse morphology and function.


Subject(s)
Lipoylation , Neurons/metabolism , Proteomics , Synapses/metabolism , Alternative Splicing/genetics , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Dendrites/metabolism , Models, Neurological , Organ Specificity , Proteome/metabolism , Rats , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
10.
Nat Neurosci ; 9(6): 824-31, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16699508

ABSTRACT

Post-translational modification by the lipid palmitate is crucial for the correct targeting and function of many proteins. Here we show that huntingtin (htt) is normally palmitoylated at cysteine 214, which is essential for its trafficking and function. The palmitoylation and distribution of htt are regulated by the palmitoyl transferase huntingtin interacting protein 14 (HIP14). Expansion of the polyglutamine tract of htt, which causes Huntington disease, results in reduced interaction between mutant htt and HIP14 and consequently in a marked reduction in palmitoylation. Mutation of the palmitoylation site of htt, making it palmitoylation resistant, accelerates inclusion formation and increases neuronal toxicity. Downregulation of HIP14 in mouse neurons expressing wild-type and mutant htt increases inclusion formation, whereas overexpression of HIP14 substantially reduces inclusions. These results suggest that the expansion of the polyglutamine tract in htt results in decreased palmitoylation, which contributes to the formation of inclusion bodies and enhanced neuronal toxicity.


Subject(s)
Carrier Proteins/metabolism , Cerebral Cortex/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Palmitic Acid/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing , Amino Acid Sequence/physiology , Animals , Animals, Newborn , COS Cells , Carrier Proteins/genetics , Cells, Cultured , Cerebral Cortex/cytology , Chlorocebus aethiops , Cysteine/metabolism , Down-Regulation/genetics , Humans , Huntingtin Protein , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Peptides/metabolism , Protein Processing, Post-Translational/physiology , Protein Transport/physiology , Rats , Trinucleotide Repeat Expansion/genetics
11.
J Biol Chem ; 281(6): 3669-78, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16338934

ABSTRACT

Myosin V motors mediate cargo transport; however, the identity of neuronal molecules transported by these proteins remains unknown. Here we show that myosin Vb is expressed in several neuronal populations and associates with the alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate-type glutamate receptor subunit GluR1. In developing hippocampal neurons, expression of the tail domain of myosin Vb, but not myosin Va, enhanced GluR1 accumulation in the soma and reduced its surface expression. These changes were accompanied by reduced GluR1 clustering and diminished frequency of excitatory but not inhibitory synaptic currents. Similar effects were observed upon expression of full-length myosin Vb lacking a C-terminal region required for binding to the small GTPase Rab11. In contrast, mutant myosin Vb did not change the localization of several other neurotransmitter receptors, including the glutamate receptor subunit NR1. These results reveal a novel mechanism for the transport of a specific glutamate receptor subunit in neurons mediated by a member of the myosin V family.


Subject(s)
Myosins/chemistry , Receptors, Glutamate/chemistry , Animals , Blotting, Western , Brain/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary/metabolism , Electrophysiology , Female , Glutathione Transferase/metabolism , Hippocampus/embryology , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Fluorescence , Mutagenesis , Mutation , Myosin Type V/chemistry , Neurons/metabolism , Neurotransmitter Agents/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Transport , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Subcellular Fractions , Transfection , rab GTP-Binding Proteins/metabolism
12.
J Biol Chem ; 280(17): 17312-9, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15723836

ABSTRACT

The balance between excitatory and inhibitory synapses is a tightly regulated process that requires differential recruitment of proteins that dictate the specificity of newly formed contacts. However, factors that control this process remain unidentified. Here we show that members of the neuroligin (NLG) family, including NLG1, NLG2, and NLG3, drive the formation of both excitatory and inhibitory presynaptic contacts. The enrichment of endogenous NLG1 at excitatory contacts and NLG2 at inhibitory synapses supports an important in vivo role for these proteins in the development of both types of contacts. Immunocytochemical and electrophysiological analysis showed that the effects on excitatory and inhibitory synapses can be blocked by treatment with a fusion protein containing the extracellular domain of neurexin-1beta. We also found that overexpression of PSD-95, a postsynaptic binding partner of NLGs, resulted in a shift in the distribution of NLG2 from inhibitory to excitatory synapses. These findings reveal a critical role for NLGs and their synaptic partners in controlling the number of inhibitory and excitatory synapses. Furthermore, relative levels of PSD-95 alter the ratio of excitatory to inhibitory synaptic contacts by sequestering members of the NLG family to excitatory synapses.


Subject(s)
Membrane Proteins/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Synapses/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules, Neuronal , Cells, Cultured , Cloning, Molecular , DNA, Complementary/metabolism , Disks Large Homolog 4 Protein , Electrophysiology , Gene Library , Green Fluorescent Proteins/metabolism , Guanylate Kinases , Hippocampus/cytology , Hippocampus/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Multigene Family , Nerve Tissue Proteins/chemistry , Neurons/metabolism , Protein Binding , Rats , Rats, Wistar , Recombinant Fusion Proteins/chemistry , Transfection
13.
Neuron ; 44(6): 977-86, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15603740

ABSTRACT

In neurons, posttranslational modification by palmitate regulates the trafficking and function of signaling molecules, neurotransmitter receptors, and associated synaptic scaffolding proteins. However, the enzymatic machinery involved in protein palmitoylation has remained elusive. Here, using biochemical assays, we show that huntingtin (htt) interacting protein, HIP14, is a neuronal palmitoyl transferase (PAT). HIP14 shows remarkable substrate specificity for neuronal proteins, including SNAP-25, PSD-95, GAD65, synaptotagmin I, and htt. Conversely, HIP14 is catalytically invariant toward paralemmin and synaptotagmin VII. Exogenous HIP14 enhances palmitoylation-dependent vesicular trafficking of several acylated proteins in both heterologous cells and neurons. Moreover, interference with endogenous expression of HIP14 reduces clustering of PSD-95 and GAD65 in neurons. These findings define HIP14 as a mammalian palmitoyl transferase involved in the palmitoylation and trafficking of multiple neuronal proteins.


Subject(s)
Carnitine O-Palmitoyltransferase/physiology , Carrier Proteins/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Palmitic Acid/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , COS Cells , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Humans , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Protein Transport/physiology , Substrate Specificity
14.
J Biol Chem ; 279(48): 50524-36, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15355980

ABSTRACT

Protein palmitoylation plays a critical role in sorting and targeting of several proteins to pre- and postsynaptic sites. In this study, we have analyzed the role of palmitoylation in trafficking of synaptotagmin I and its modulation by synaptic activity. We found that palmitoylation of N-terminal cysteines contributed to sorting of synaptotagmin I to an intracellular vesicular compartment at the presynaptic terminal. Presynaptic targeting is a unique feature of N-terminal sequences of synaptotagmin I because the palmitoylated N terminus of synaptotagmin VII failed to localize to presynaptic sites. We also found that palmitate was stably associated with both synaptotagmin I and SNAP-25 and that rapid neuronal depolarization did not affect palmitate turnover on these proteins. However, long-term treatment with drugs that either block synaptic activity or disrupt SNARE complex assembly modulated palmitoylation and accumulation of synaptotagmin I at presynaptic sites. We conclude that palmitoylation is involved in trafficking of specific elements involved in transmitter release and that distinct mechanisms regulate addition and removal of palmitate on select neuronal proteins.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Palmitates/metabolism , Synapses/metabolism , Animals , Endocytosis/physiology , Humans , Membrane Proteins/metabolism , Protein Transport/physiology , Rats , Synaptosomal-Associated Protein 25 , Synaptotagmin I , Synaptotagmins
15.
J Cell Sci ; 116(Pt 15): 3213-9, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12829740

ABSTRACT

Postsynaptic density protein 95 (PSD-95/SAP-90) is a palmitoylated membrane-associated guanylate kinase that oligomerizes and clusters ion channels and associated signaling machinery at excitatory synapses in brain. However, the mechanism for PSD-95 oligomerization and its relationship to ion channel clustering remain uncertain. Here, we find that multimerization of PSD-95 is determined by only its first 13 amino acids, which also have a remarkable capacity to oligomerize heterologous proteins. Multimerization does not involve a covalent linkage but rather palmitoylation of two cysteine residues in the 13 amino acid motif. This lipid-mediated oligomerization is a specific property of the PSD-95 motif, because it is not observed with other palmitoylated domains. Clustering K+ channel Kv1.4 requires interaction of palmitoylated PSD-95 with tetrameric K+ channel subunits but, surprisingly, does not require multimerization of PSD-95. Finally, disrupting palmitoylation with 2-bromopalmitate disperses PSD-95/K+-channel clusters. These data suggest new models for K+ channel clustering by PSD-95 - a reversible process regulated by protein palmitoylation.


Subject(s)
Lipid Metabolism , Nerve Tissue Proteins/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular , Fluorescent Antibody Technique, Indirect , Humans , Nerve Tissue Proteins/physiology , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Protein Subunits/metabolism , Protein Subunits/physiology
16.
J Cell Sci ; 115(Pt 18): 3675-82, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12186953

ABSTRACT

The Dictyostelium genome contains a single rapA gene, which encodes a Rap1 monomeric G protein. As attempts at generating rapA-null Dictyostelium cells had been unsuccessful, expression of antisense RNA from the rapA gene under control of the folate repressible discoidin promoter was used to reduce cellular levels of the Rap1 protein. As Rap1 levels gradually decreased following antisense rapA RNA induction, growth rate and cell viability also decreased, a result consistent with the idea that rapA is an essential gene. The Rap1-depleted cells exhibited reduced viability in response to osmotic shock. The accumulation of cGMP in response to 0.4 M sorbitol was reduced after rapA antisense RNA induction and was enhanced in cells expressing the constitutively activated Rap1(G12V) protein, suggesting a role for Rap1 in the generation of cGMP. Dictyostelium Rap1 formed a complex with the Ras-binding domain of RalGDS only when it was in a GTP-bound state. This assay was used to demonstrate that activation of Rap1 in response to 0.4 M sorbitol occurred with initial kinetics similar to those observed for the accumulation of cGMP. Furthermore, the addition of 2 mM EDTA to osmotically shocked cells, a treatment that enhances cGMP accumulation, also enhanced Rap1 activation. These results suggest a direct role for Rap1 in the activation of guanylyl cyclase during the response to hyperosmotic conditions. Rap1 was also activated in response to low temperature but not in response to low osmolarity or high temperature.


Subject(s)
Cell Survival/genetics , Cyclic GMP/metabolism , Dictyostelium/metabolism , Osmotic Pressure , Stress, Physiological/metabolism , rap1 GTP-Binding Proteins/deficiency , Animals , Cell Division/genetics , Cell Size/genetics , Dictyostelium/genetics , Edetic Acid/pharmacology , Gene Expression Regulation/genetics , Guanylate Cyclase/metabolism , Osmotic Pressure/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , RNA, Antisense/genetics , Sorbitol/pharmacology , Stress, Physiological/genetics , Temperature , ral Guanine Nucleotide Exchange Factor/genetics , ral Guanine Nucleotide Exchange Factor/metabolism , rap1 GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...