Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 276(11): 7705-8, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11244087

ABSTRACT

Phosphatidylinositolpolyphosphates (PIPs) are centrally involved in many biological processes, ranging from cell growth and organization of the actin cytoskeleton to endo- and exocytosis. Phosphorylation of phosphatidylinositol at the D-4 position, an essential step in the biosynthesis of PIPs, appears to be catalyzed by two biochemically distinct enzymes. However, only one of these two enzymes has been molecularly characterized. We now describe a novel class of phosphatidylinositol 4-kinases that probably corresponds to the missing element in phosphatidylinositol metabolism. These kinases are highly conserved evolutionarily, but unrelated to previously characterized phosphatidylinositol kinases, and thus represent the founding members of a new family. The novel phosphatidylinositol 4-kinases, which are widely expressed in cells, only phosphorylate phosphatidylinositol, are potently inhibited by adenosine, but are insensitive to wortmannin or phenylarsine oxide. Although they lack an obvious transmembrane domain, they are strongly attached to membranes by palmitoylation. Our data suggest that independent pathways for phosphatidylinositol 4-phosphate synthesis emerged during evolution, possibly to allow tight temporal and spatial control over the production of this key signaling molecule.


Subject(s)
1-Phosphatidylinositol 4-Kinase/chemistry , Yeasts/enzymology , Amino Acid Sequence , Animals , COS Cells , Cattle , Conserved Sequence , Humans , Molecular Sequence Data , Molecular Weight , Phylogeny , Rats
2.
Neuroscience ; 101(3): 793-802, 2000.
Article in English | MEDLINE | ID: mdl-11113328

ABSTRACT

At the synapse, neurotransmitter release is triggered physiologically by Ca(2+) influx through voltage-gated Ca(2+) channels. Non-physiologically, release can be evoked by a potent neurotoxin, alpha-latrotoxin, and by hypertonic sucrose. Controversy has arisen on whether release evoked by alpha-latrotoxin and hypertonic sucrose requires extracellular Ca(2+) or Ca(2+) from intracellular stores. Using synaptosomes, we have studied the Ca(2+) dependence of alpha-latrotoxin and sucrose action in different neurotransmitter systems. In agreement with previous data, no requirement for extracellular Ca(2+) in sucrose-induced secretion of norepinephrine, dopamine, glutamate or GABA was detected. Unexpectedly, we observed large differences between these neurotransmitters in the Ca(2+) dependence of alpha-latrotoxin-stimulated release: norepinephrine release required Ca(2+), dopamine release was only partially Ca(2+) dependent, and glutamate and GABA release did not require Ca(2+). To test if Ca(2+) derived from intracellular Ca(2+) stores participates in neurotransmitter release triggered by alpha-latrotoxin or hypertonic sucrose, we employed thapsigargin, a Ca(2+)-ATPase inhibitor that empties Ca(2+) stores. Thapsigargin did not induce neurotransmitter release, nor did it inhibit subsequent release stimulated by KCl depolarization, hypertonic sucrose or alpha-latrotoxin. However, intracellular Ca(2+) performs an important regulatory function, since thapsigargin increased the size of the readily releasable pool as measured by stimulation with hypertonic sucrose. This effect required extracellular Ca(2+) and protein kinase C, suggesting that depletion of internal Ca(2+) stores leads to store-operated Ca(2+) entry. The resulting Ca(2+) influx does not trigger release by itself, but activates protein kinase C that increases the readily releasable pool of neurotransmitters. Our data show that internal and external Ca(2+) is not acutely involved in hypertonic sucrose-evoked neurotransmitter release, while alpha-latrotoxin-triggered release requires external Ca(2+) for a subset of neurotransmitters. Although internal Ca(2+) is not essential for release, it modulates its extent, implying that the emptying of intracellular stores by activation of presynaptic receptors plays an important regulatory role in neurotransmitter release.


Subject(s)
Neurotransmitter Agents/metabolism , Spider Venoms/pharmacology , Sucrose/pharmacology , Synaptic Transmission/drug effects , Synaptic Vesicles/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/physiology , Glutamic Acid/metabolism , Hypertonic Solutions/pharmacology , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Male , Mice , Mice, Inbred C57BL , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology , Synaptosomes/metabolism , Thapsigargin/pharmacology , gamma-Aminobutyric Acid/metabolism
3.
EMBO J ; 19(13): 3250-62, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880438

ABSTRACT

alpha-latrotoxin, a component of black widow spider venom, binds to presynaptic nerve terminals and stimulates massive neurotransmitter release. Previous studies have demonstrated that alpha-latrotoxin first binds to two high-affinity receptors on nerve terminals, neurexins and CLs (CIRLs and latrophilins), and then executes a critical, second step of unknown nature that stimulates neurotransmitter release. We now demonstrate that incubation of alpha-latrotoxin with synaptosomes at 0 degrees C results in its peripheral membrane association. Incubation at 37 degrees C, however, converts the toxin into an operationally integral membrane protein, and induces generation of a protease-resistant fragment that consists of the entire N-terminal domain of alpha-latrotoxin and becomes protease sensitive after lysis of synaptosomes. Our data suggest that alpha-latrotoxin inserts into the presynaptic plasma membrane after receptor binding, resulting in an intracellular location of the N-terminal sequences. Membrane insertion of the N-terminal domain of alpha-latrotoxin occurs spontaneously, independently of membrane recycling or transmembrane ion gradients. We postulate that alpha-latrotoxin acts intracellularly in triggering release, and propose that non-selective cation channels induced by alpha-latrotoxin may be a by-product of membrane insertion.


Subject(s)
Presynaptic Terminals/drug effects , Spider Venoms/pharmacology , Animals , Biological Transport , Cell Membrane/drug effects , Hot Temperature , Hydrolysis , Mice , Peptide Fragments/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spider Venoms/chemistry , Spider Venoms/metabolism , Synaptosomes/drug effects , Trypsin/metabolism
4.
EMBO J ; 17(21): 6188-99, 1998 Nov 02.
Article in English | MEDLINE | ID: mdl-9799228

ABSTRACT

alpha-Latrotoxin stimulates neurotransmitter release probably by binding to two receptors, CIRL/latrophilin 1 (CL1) and neurexin Ialpha. We have now produced recombinant alpha-latrotoxin (LtxWT) that is as active as native alpha-latrotoxin in triggering synaptic release of glutamate, GABA and norepinephrine. We have also generated three alpha-latrotoxin mutants with substitutions in conserved cysteine residues, and a fourth mutant with a four-residue insertion. All four alpha-latrotoxin mutants were found to be unable to trigger release. Interestingly, the insertion mutant LtxN4C exhibited receptor-binding affinities identical to wild-type LtxWT, bound to CL1 and neurexin Ialpha as well as LtxWT, and similarly stimulated synaptic hydrolysis of phosphatidylinositolphosphates. Therefore, receptor binding by alpha-latrotoxin and stimulation of phospholipase C are insufficient to trigger exocytosis. This conclusion was confirmed in experiments with La3+ and Cd2+. La3+ blocked release triggered by LtxWT, whereas Cd2+ enhanced it. Both cations, however, had no effect on the stimulation by LtxWT of phosphatidylinositolphosphate hydrolysis. Our data show that receptor binding by alpha-latrotoxin and activation of phospholipase C do not by themselves trigger exocytosis. Thus receptors recruit alpha-latrotoxin to its point of action without activating exocytosis. Exocytosis probably requires an additional receptor-independent activity of alpha-latrotoxin that is selectively inhibited by the LtxN4C mutation and by La3+.


Subject(s)
Exocytosis/physiology , Receptors, Peptide/metabolism , Signal Transduction/physiology , Spider Venoms/metabolism , Animals , Cadmium/pharmacology , Cysteine/metabolism , Glycoproteins , Lanthanum/pharmacology , Mice , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neuropeptides , Neurotransmitter Agents/metabolism , Phosphatidylinositols/metabolism , Protein Binding/genetics , Recombinant Proteins/genetics , Spider Venoms/genetics , Synaptosomes/metabolism , Type C Phospholipases/metabolism
5.
J Biol Chem ; 273(49): 32715-24, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9830014

ABSTRACT

alpha-Latrotoxin, a potent excitatory neurotoxin, binds to two receptors: a G-protein-coupled receptor called CIRL/latrophilin 1 (CL1) and a cell-surface protein called neurexin Ialpha. We now show that CL1 belongs to a family of closely related receptors called CL1, CL2, and CL3. CLs exhibit an unusual multidomain structure with similar alternative splicing and large extra- and intracellular sequences. CLs share domains with other G-protein-coupled receptors, lectins, and olfactomedins/myocilin. In addition, CLs contain a novel, widespread cysteine-rich domain that may direct endoproteolytic processing of CLs during transport to the cell surface. Although the mRNAs for CLs are enriched in brain, CLs are ubiquitously expressed in all tissues. To examine how binding of alpha-latrotoxin to CL1 triggers exocytosis, we used PC12 cells transfected with human growth hormone. Ca2+-dependent secretion of human growth hormone from transfected PC12 cells was triggered by KCl depolarization or alpha-latrotoxin and was inhibited by tetanus toxin and by phenylarsine oxide, a phosphoinositide kinase inhibitor. When CL1 was transfected into PC12 cells, their response to alpha-latrotoxin was sensitized dramatically. A similar sensitization to alpha-latrotoxin was observed with different splice variants of CL1, whereas CL2 and CL3 were inactive in this assay. A truncated form of CL1 that contains only a single transmembrane region and presumably is unable to mediate G-protein-signaling was as active as wild type CL1 in alpha-latrotoxin-triggered exocytosis. Our data show that CL1, CL2, and CL3 perform a general and ubiquitous function as G-protein-coupled receptors in cellular signaling. In addition, CL1 serves a specialized role as an alpha-latrotoxin receptor that does not require G-protein-signaling for triggering exocytosis. This suggests that as an alpha-latrotoxin receptor, CL1 recruits alpha-latrotoxin to target membranes without participating in exocytosis directly.


Subject(s)
Exocytosis , GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Peptide/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Exocytosis/drug effects , Humans , Molecular Sequence Data , PC12 Cells , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Sequence Homology, Amino Acid , Spider Venoms/pharmacology , Structure-Activity Relationship , Transfection
6.
J Biol Chem ; 273(34): 21451-4, 1998 Aug 21.
Article in English | MEDLINE | ID: mdl-9705272

ABSTRACT

Newly synthesized phosphatidylinositol phosphates have been implicated in many membrane-trafficking reactions. They are essential for exocytosis of norepinephrine in PC12 cells and chromaffin cells, suggesting a function in membrane fusion. We have now studied the role of phosphatidylinositol phosphates in synaptic vesicle exocytosis using synaptosomes. Under conditions where phosphorylation of phosphatidylinositols is blocked, norepinephrine secretion was nearly abolished whereas glutamate and GABA release was still elicited. Thus phosphatidylinositides are essential only for some membrane fusion reactions, and exocytotic release mechanisms differ between neurotransmitters.


Subject(s)
Norepinephrine/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Arsenicals/pharmacology , Calcium/metabolism , Chromaffin Cells/metabolism , Enzyme Inhibitors/pharmacology , Exocytosis , Glutamic Acid/metabolism , Neurotransmitter Agents/metabolism , PC12 Cells , Phosphatidylinositol 4,5-Diphosphate/biosynthesis , Rats , Synaptosomes/drug effects
7.
J Biol Chem ; 273(4): 2306-11, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9442075

ABSTRACT

Phosphatidylinositol phosphates (PIPs) perform central functions in signal transduction and membrane traffic. Synaptojanin is a PIP 5-phosphatase that is expressed in a brain-specific and a ubiquitous splice variants and is thought to constitute the major PIP 5-phosphatase in mammalian brain (Woscholski, R., Finan, P.M., Radley, E., Totty, N.F., Sterling, A.E., Hsuan, J.J., Waterfield, M. D., and Parker, P. J. (1997) J. Biol. Chem. 272, 9625-9628). We now describe synaptojanin 2, a novel isoform of synaptojanin that, similar to synaptojanin 1, contains an N-terminal SAC1-like sequence and a central 5-phosphatase domain but a distinct, unique C-terminal sequence. Transfection studies demonstrated that synaptojanin 2, like synaptojanin 1, is an active PIP phosphatase. An interesting feature of synaptojanin 1 is the presence of a long open reading frame in the 3' region of the brain mRNA that in non-brain tissues is joined to the coding region by alternative splicing, resulting in a shorter synaptojanin 1 form in brain and a longer form in peripheral tissues (Ramjaun, A. R., and McPherson, P. S. (1996) J. Biol. Chem. 271, 24856-24861). Although it exhibits no homology to synaptojanin 1 in this region, synaptojanin 2 also contains an open reading frame in the 3' region that is subject to alternative splicing. Similar to synaptojanin 1, alternative splicing of synaptojanin 2 is tissue-specific and creates a shorter isoform expressed in brain and a longer form in peripheral tissues. The similar alternative splicing of two homologous proteins in a region of non-homology raises the possibility of evolutionary convergence and supports the significance of the variants. Analysis of mRNAs from three brain regions at different developmental stages revealed that alternative splicing of synaptojanin 2 is a developmentally late event, occurring only after the first postnatal week after the generation of neurons and initial synaptogenesis.


Subject(s)
Alternative Splicing , Nerve Tissue Proteins/genetics , Phosphoric Monoester Hydrolases/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Cloning, Molecular , Enzyme Inhibitors/chemistry , Gene Library , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Phosphatidylinositol Phosphates/physiology , Phospholipase D/antagonists & inhibitors , Phosphoric Monoester Hydrolases/chemistry , Signal Transduction
8.
J Biol Chem ; 273(3): 1705-10, 1998 Jan 16.
Article in English | MEDLINE | ID: mdl-9430716

ABSTRACT

alpha-Latrotoxin is a potent neurotoxin from black widow spider venom that binds to presynaptic receptors and causes massive neurotransmitter release. A surprising finding was the biochemical description of two distinct cell surface proteins that bind alpha-latrotoxin with nanomolar affinities; Neurexin I alpha binds alpha-latrotoxin in a Ca(2+)-dependent manner, and CIRL/latrophilin binds in a Ca(2+)-independent manner. We have now generated and analyzed mice that lack neurexin I alpha to test its importance in alpha-latrotoxin action. alpha-Latrotoxin binding to brain membranes from mutant mice was decreased by almost 50% compared with wild type membranes; the decrease was almost entirely due to a loss of Ca(2+)-dependent alpha-latrotoxin binding sites. In cultured hippocampal neurons, alpha-latrotoxin was still capable of activating neurotransmission in the absence of neurexin I alpha. Direct measurements of [3H]glutamate release from synaptosomes, however, showed a major decrease in the amount of release triggered by alpha-latrotoxin in the presence of Ca2+. Thus neurexin I alpha is not essential for alpha-latrotoxin action but contributes to alpha-latrotoxin action when Ca2+ is present. Viewed as a whole, our results show that mice contain two distinct types of alpha-latrotoxin receptors with similar affinities and abundance but different properties and functions. The action of alpha-latrotoxin may therefore be mediated by independent parallel pathways, of which the CIRL/latrophilin pathway is sufficient for neurotransmitter release, whereas the neurexin I alpha pathway contributes to the Ca(2+)-dependent action of alpha-latrotoxin.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, Peptide/metabolism , Spider Venoms/metabolism , Alternative Splicing , Animals , Brain/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Chromosome Mapping , Glutamic Acid/metabolism , Glycoproteins , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neuropeptides , Receptors, Peptide/genetics , Spider Venoms/genetics , Synaptic Transmission , Synaptosomes/metabolism
9.
J Biol Chem ; 271(13): 7535-43, 1996 Mar 29.
Article in English | MEDLINE | ID: mdl-8631785

ABSTRACT

The venom of the black widow spider (BWSV) (Latrodectus mactans tredecimguttatus) contains several potent, high molecular mass (>110 kDa) neurotoxins that cause neurotransmitter release in a phylum-specific manner. The molecular mechanism of action of these proteins is poorly understood because their structures are largely unknown, and they have not been functionally expressed. This study reports on the primary structure of delta-latroinsectotoxin (delta-LIT), a novel insect-specific toxin from BWSV, that contains 1214 amino acids. delta-LIT comprises four structural domains: a signal peptide followed by an N-terminal domain that exhibits the highest degree of identity with other latrotoxins, a central region composed of 15 ankyrin-like repeats, and a C-terminal domain. The domain organization of delta-LIT is similar to that of other latrotoxins, suggesting that these toxins are a family of related proteins. The predicted molecular mass and apparent mobility of the protein (approximately 130 kDa) encoded in the delta-LIT gene differs from that of native delta-LIT purified from BWSV (approximately 100 kDa), suggesting that the toxin is produced by proteolytic processing of a precursor. MALDI-MS of purified native delta-LIT revealed a molecular ion with m/z+ of 110916 +/- 100, indicating that the native delta-LIT is 991 amino acids in length. When the full-length delta-LIT cDNA was expressed in bacteria the protein product was inactive, but expression of a C-terminally truncated protein containing 991 residues produced a protein that caused massive neurotransmitter release at the locust neuromuscular junction at nanomolar concentrations. Channels formed in locust muscle membrane and artificial lipid bilayers by the native delta-LIT have a high Ca2+ permeability, whereas those formed by truncated, recombinant protein do not.


Subject(s)
Black Widow Spider/metabolism , Gene Expression , Spider Venoms/biosynthesis , Spider Venoms/chemistry , Amino Acid Sequence , Animals , Ankyrins/chemistry , Base Sequence , Cloning, Molecular , Consensus Sequence , DNA Primers , DNA, Complementary , Escherichia coli , Insecta , Lipid Bilayers , Mass Spectrometry , Membrane Potentials/drug effects , Molecular Sequence Data , Molecular Weight , Muscles/drug effects , Muscles/physiology , Oligonucleotide Probes , Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Deletion , Sequence Homology, Amino Acid , Spider Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL