Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Mol Cell ; 50(2): 172-84, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23499007

ABSTRACT

The Uba6 (E1)-Use1 (E2) ubiquitin transfer cascade is a poorly understood alternative arm of the ubiquitin proteasome system (UPS) and is required for mouse embryonic development, independent of the canonical Uba1-E2-E3 pathway. Loss of neuronal Uba6 during embryonic development results in altered patterning of neurons in the hippocampus and the amygdala, decreased dendritic spine density, and numerous behavioral disorders. The levels of the E3 ubiquitin ligase Ube3a (E6-AP) and Shank3, both linked with dendritic spine function, are elevated in the amygdala of Uba6-deficient mice, while levels of the Ube3a substrate Arc are reduced. Uba6 and Use1 promote proteasomal turnover of Ube3a in mouse embryo fibroblasts (MEFs) and catalyze Ube3a ubiquitylation in vitro. These activities occur in parallel with an independent pathway involving Uba1-UbcH7, but in a spatially distinct manner in MEFs. These data reveal an unanticipated role for Uba6 in neuronal development, spine architecture, mouse behavior, and turnover of Ube3a.


Subject(s)
Amygdala/abnormalities , CA3 Region, Hippocampal/abnormalities , Qc-SNARE Proteins/deficiency , Ubiquitin-Activating Enzymes/deficiency , Ubiquitination , Amygdala/enzymology , Amygdala/pathology , Animals , Body Weight , CA3 Region, Hippocampal/enzymology , CA3 Region, Hippocampal/pathology , Cells, Cultured , Dendritic Spines/pathology , Embryonic Development , Energy Metabolism , Female , Genes, Lethal , Learning Disabilities/metabolism , Locomotion , Male , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/metabolism , Oxygen Consumption , Protein Stability , Protein Structure, Tertiary , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , SNARE Proteins , Social Behavior , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins
2.
PLoS Pathog ; 14(5): e1007028, 2018 05.
Article in English | MEDLINE | ID: mdl-29746582

ABSTRACT

Positive-strand RNA viruses assemble numerous membrane-bound viral replicase complexes within large replication compartments to support their replication in infected cells. Yet the detailed mechanism of how given subcellular compartments are subverted by viruses is incompletely understood. Although, Tomato bushy stunt virus (TBSV) uses peroxisomal membranes for replication, in this paper, we show evidence that the ER-resident SNARE (soluble NSF attachment protein receptor) proteins play critical roles in the formation of active replicase complexes in yeast model host and in plants. Depletion of the syntaxin 18-like Ufe1 and Use1, which are components of the ER SNARE complex in the ERAS (ER arrival site) subdomain, in yeast resulted in greatly reduced tombusvirus accumulation. Over-expression of a dominant-negative mutant of either the yeast Ufe1 or the orthologous plant Syp81 syntaxin greatly interferes with tombusvirus replication in yeast and plants, thus further supporting the role of this host protein in tombusvirus replication. Moreover, tombusvirus RNA replication was low in cell-free extracts from yeast with repressed Ufe1 or Use1 expression. We also present evidence for the mislocalization of the tombusviral p33 replication protein to the ER membrane in Ufe1p-depleted yeast cells. The viral p33 replication protein interacts with both Ufe1p and Use1p and co-opts them into the TBSV replication compartment in yeast and plant cells. The co-opted Ufe1 affects the virus-driven membrane contact site formation, sterol-enrichment at replication sites, recruitment of several pro-viral host factors and subversion of the Rab5-positive PE-rich endosomes needed for robust TBSV replication. In summary, we demonstrate a critical role for Ufe1 and Use1 SNARE proteins in TBSV replication and propose that the pro-viral functions of Ufe1 and Use1 are to serve as assembly hubs for the formation of the extensive TBSV replication compartments in cells. Altogether, these findings point clearly at the ERAS subdomain of ER as a critical site for the biogenesis of the TBSV replication compartment.


Subject(s)
SNARE Proteins/metabolism , SNARE Proteins/physiology , Tombusvirus/physiology , DNA Replication , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Endosomes/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Mitochondrial Membranes/metabolism , Qa-SNARE Proteins/metabolism , Qa-SNARE Proteins/physiology , Qc-SNARE Proteins/metabolism , Qc-SNARE Proteins/physiology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Tombusvirus/genetics , Tombusvirus/metabolism , Tombusvirus/pathogenicity , Viral Proteins/genetics , Virus Replication/physiology
3.
FASEB J ; 33(5): 6023-6034, 2019 05.
Article in English | MEDLINE | ID: mdl-30742775

ABSTRACT

Hepatitis B virus (HBV) replication and envelopment is dependent on cellular autophagy. Previously, we have provided evidence for the extensive lysosomal degradation of HBV virions and the hepatitis B surface antigen (HBsAg), which is likely controlled by autophagosome-lysosome fusion. Synaptosomal-associated protein 29 (SNAP29) has been identified as a protein specifically mediating autophagosome-lysosome fusion. Thus, in the present study, we addressed the hypothesis that SNAP29 is required for the autophagic degradation of HBV virions and HBsAg. We found that silencing SNAP29 significantly increased the number of autophagosomes and concomitantly promoted HBV replication and HBsAg production. Conversely, SNAP29 overexpression decreased HBV production. Consistent with this, SNAP29 modulated HBV production by interacting with vesicle-associated membrane protein 8 (VAMP8) and synergistically regulated HBV replication with Rab7 complexes. Moreover, the production and release of the small HBsAg is strongly regulated by SNAP29 expression, suggesting that its export occurs partly through the autophagic pathway. Our findings provide new evidence, strongly suggesting that autophagic degradation critically determines the production of HBV virions and HBsAg and that this is controlled by the SNAP29-VAMP8 interaction.-Lin, Y., Wu, C., Wang, X., Liu, S., Kemper, T., Li, F., Squire, A., Zhu, Y., Zhang, J., Chen, X., Lu, M. Synaptosomal-associated protein 29 is required for the autophagic degradation of hepatitis B virus.


Subject(s)
Autophagy , Hepatitis B Surface Antigens/metabolism , Hepatitis B/metabolism , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , R-SNARE Proteins/metabolism , Synaptosomes/metabolism , Animals , Autophagosomes/metabolism , Cattle , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Viral , Gene Silencing , Hep G2 Cells , Hepatitis B/virology , Hepatitis B virus , Humans , Lysosomes/metabolism , Membrane Fusion , RNA, Small Interfering/metabolism , Serum Albumin, Bovine/metabolism , Virion , Virus Replication
4.
J Cell Sci ; 127(Pt 8): 1712-25, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24496451

ABSTRACT

Acquisition of an invasive phenotype is prerequisite for tumor metastasis. Degradation of the extracellular matrix (ECM), and subsequent invasion by tumor cells, is mediated, in part, through subcellular structures called invadopodia. Src-dependent cytoskeletal rearrangements are required to form invadopodia, and here we identify an association between Src, epidermal growth factor receptor (EGFR), and ß1 integrin that facilitates invadopodia formation. The association of Src, EGFR and ß1 integrin is dependent upon membrane traffic that is mediated by syntaxin13 (officially known as STX12) and SNAP23; a similar dependence on these two SNARE proteins was observed for invadopodium-based matrix degradation and cell invasion. Inhibition of SNARE function impaired the delivery of Src and EGFR to developing invadopodia, as well as the ß1-integrin-dependent activation of Src and phosphorylation of EGFR on Tyr residue 845. We also identified an association between SNAP23 and ß1 integrin, and inhibition of ß1 integrin increased this association, whereas the interaction between syntaxin13 and SNAP23 was reduced. The results suggest that SNARE-dependent trafficking is regulated, in part, by ß1 integrin and is required for the delivery of Src and EGFR to sites of invadopodia formation in order to support tumor cell invasion.


Subject(s)
ErbB Receptors/metabolism , Integrin beta1/metabolism , Qa-SNARE Proteins/physiology , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Pseudopodia/metabolism
5.
EMBO J ; 27(15): 2031-42, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18650938

ABSTRACT

The homotypic fusion of yeast vacuoles, each with 3Q- and 1R-SNARE, requires SNARE chaperones (Sec17p/Sec18p and HOPS) and regulatory lipids (sterol, diacylglycerol and phosphoinositides). Pairs of liposomes of phosphatidylcholine/phosphatidylserine, bearing three vacuolar Q-SNAREs on one and the R-SNARE on the other, undergo slow lipid mixing, but this is unaffected by HOPS and inhibited by Sec17p/Sec18p. To study these essential fusion components, we reconstituted proteoliposomes of a more physiological composition, bearing vacuolar lipids and all four vacuolar SNAREs. Their fusion requires Sec17p/Sec18p and HOPS, and each regulatory lipid is important for rapid fusion. Although SNAREs can cause both fusion and lysis, fusion of these proteoliposomes with Sec17p/Sec18p and HOPS is not accompanied by lysis. Sec17p/Sec18p, which disassemble SNARE complexes, and HOPS, which promotes and proofreads SNARE assembly, act synergistically to form fusion-competent SNARE complexes, and this synergy requires phosphoinositides. This is the first chemically defined model of the physiological interactions of these conserved fusion catalysts.


Subject(s)
Adenosine Triphosphatases/metabolism , Lipids/physiology , Membrane Fusion/physiology , Molecular Chaperones/physiology , SNARE Proteins/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adenosine Triphosphatases/chemistry , Genes, Fungal , Lipids/chemistry , Liposomes , Molecular Chaperones/chemistry , Phosphatidylcholines/chemistry , Phosphatidylserines/chemistry , Protein Binding , Protein Transport , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/physiology , Qb-SNARE Proteins/chemistry , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/chemistry , Qc-SNARE Proteins/physiology , R-SNARE Proteins/chemistry , R-SNARE Proteins/physiology , Saccharomyces cerevisiae Proteins/chemistry , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Vesicular Transport Proteins/chemistry
6.
J Biol Chem ; 285(32): 24629-38, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20519516

ABSTRACT

SNAP25 plays an essential role in neuronal exocytosis pathways. SNAP25a and SNAP25b are alternatively spliced isoforms differing by only nine amino acids, three of which occur within the palmitoylated cysteine-rich domain. SNAP23 is 60% identical to SNAP25 and has a distinct cysteine-rich domain to both SNAP25a and SNAP25b. Despite the conspicuous differences within the palmitoylated domains of these secretory proteins, there is no information on their comparative interactions with palmitoyl transferases. We report that membrane association of all SNAP25/23 proteins is enhanced by Golgi-localized DHHC3, DHHC7, and DHHC17. In contrast, DHHC15 promoted a statistically significant increase in membrane association of only SNAP25b. To investigate the underlying cause of this differential specificity, we examined a SNAP23 point mutant (C79F) designed to mimic the cysteine-rich domain of SNAP25b. DHHC15 promoted a marked increase in membrane binding and palmitoylation of this SNAP23 mutant, demonstrating that the distinct cysteine-rich domains of SNAP25/23 contribute to differential interactions with DHHC15. The lack of activity of DHHC15 toward wild-type SNAP23 was not overcome by replacing its DHHC domain with that from DHHC3, suggesting that substrate specificity is not determined by the DHHC domain alone. Interestingly, DHHC2, which is closely related to DHHC15, associates with the plasma membrane in PC12 cells and can palmitoylate all SNAP25 isoforms. DHHC2 is, thus, a candidate enzyme to regulate SNAP25/23 palmitoylation dynamics at the plasma membrane. Finally, we demonstrate that overexpression of specific Golgi-localized DHHC proteins active against SNAP25/23 proteins perturbs the normal secretion of human growth hormone from PC12 cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Palmitic Acid/chemistry , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , Synaptosomal-Associated Protein 25/physiology , Vesicular Transport Proteins/physiology , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cysteine/chemistry , Golgi Apparatus/metabolism , Humans , Molecular Sequence Data , PC12 Cells , Protein Isoforms , Rats , Sequence Homology, Amino Acid , Synaptosomal-Associated Protein 25/metabolism
8.
Gene Expr ; 20(1): 25-37, 2020 06 12.
Article in English | MEDLINE | ID: mdl-31757226

ABSTRACT

Hepatic stellate cells (HSC) are critical effector cells of liver fibrosis. In the injured liver, HSC differentiate into a myofibrobastic phenotype. A critical feature distinguishing myofibroblastic from quiescent HSC is cytoskeletal reorganization. Soluble NSF attachment receptor (SNARE) proteins are important in trafficking of newly synthesized proteins to the plasma membrane for release into the extracellular environment. The goals of this project were to determine the expression of specific SNARE proteins in myofibroblastic HSC and to test whether their alteration changed the HSC phenotype in vitro and progression of liver fibrosis in vivo. We found that HSC lack the t-SNARE protein, SNAP-25, but express a homologous protein, SNAP-23. Downregulation of SNAP-23 in HSC induced reduction in polymerization and disorganization of the actin cytoskeleton associated with loss of cell movement. In contrast, reduction in SNAP-23 in mice by monogenic deletion delayed but did not prevent progression of liver fibrosis to cirrhosis. Taken together, these findings suggest that SNAP-23 is an important regular of actin dynamics in myofibroblastic HSC, but that the role of SNAP-23 in the progression of liver fibrosis in vivo is unclear.


Subject(s)
Actin Cytoskeleton/ultrastructure , Hepatic Stellate Cells/ultrastructure , Myofibroblasts/ultrastructure , Qb-SNARE Proteins/deficiency , Qc-SNARE Proteins/deficiency , Actin Cytoskeleton/chemistry , Actin Depolymerizing Factors/biosynthesis , Actins/analysis , Animals , Carbon Tetrachloride/toxicity , Cell Line , Cell Movement , Cell Separation , Gene Knockdown Techniques , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Qb-SNARE Proteins/antagonists & inhibitors , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/antagonists & inhibitors , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Stress Fibers/chemistry , Stress Fibers/ultrastructure , Wound Healing , rho-Associated Kinases/physiology
9.
Mol Biol Cell ; 20(20): 4435-43, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19710423

ABSTRACT

The endoplasmic reticulum (ER) is proposed to be a membrane donor for phagosome formation. In support of this, we have previously shown that the expression level of syntaxin 18, an ER-localized SNARE protein, correlates with phagocytosis activity. To obtain further insights into the involvement of the ER in phagocytosis we focused on Sec22b, another ER-localized SNARE protein that is also found on phagosomal membranes. In marked contrast to the effects of syntaxin 18, we report here that phagocytosis was nearly abolished in J774 macrophages stably expressing mVenus-tagged Sec22b, without affecting the cell surface expression of the Fc receptor or other membrane proteins related to phagocytosis. Conversely, the capacity of the parental J774 cells for phagocytosis was increased when endogenous Sec22b expression was suppressed. Domain analyses of Sec22b revealed that the R-SNARE motif, a selective domain for forming a SNARE complex with syntaxin18 and/or D12, was responsible for the inhibition of phagocytosis. These results strongly support the ER-mediated phagocytosis model and indicate that Sec22b is a negative regulator of phagocytosis in macrophages, most likely by regulating the level of free syntaxin 18 and/or D12 at the site of phagocytosis.


Subject(s)
Macrophages/physiology , Phagocytosis/physiology , Qa-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , R-SNARE Proteins/physiology , SNARE Proteins/physiology , Amino Acid Motifs , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Mice , Opsonin Proteins/metabolism , Protein Interaction Mapping , Protein Structure, Tertiary , Qa-SNARE Proteins/chemistry , Qc-SNARE Proteins/chemistry , R-SNARE Proteins/chemistry , RNA, Small Interfering/pharmacology , Receptors, IgG/metabolism , Recombinant Fusion Proteins/physiology , SNARE Proteins/chemistry , Vesicular Transport Proteins , Zymosan/metabolism
10.
PLoS One ; 4(5): e5449, 2009.
Article in English | MEDLINE | ID: mdl-19421331

ABSTRACT

BACKGROUND: Growth and division of Saccharomyces cerevisiae is dependent on the action of SNARE proteins that are required for membrane fusion. SNAREs are regulated, through a poorly understood mechanism, to ensure membrane fusion at the correct time and place within a cell. Although fusion of secretory vesicles with the plasma membrane is important for yeast cell growth, the relationship between exocytic SNAREs and cell physiology has not been established. METHODOLOGY/PRINCIPAL FINDINGS: Using genetic analysis, we identified several influences on the function of exocytic SNAREs. Genetic disruption of the V-ATPase, but not vacuolar proteolysis, can suppress two different temperature-sensitive mutations in SEC9. Suppression is unlikely due to increased SNARE complex formation because increasing SNARE complex formation, through overexpression of SRO7, does not result in suppression. We also observed suppression of sec9 mutations by growth on alkaline media or on a non-fermentable carbon source, conditions associated with a reduced growth rate of wild-type cells and decreased SNARE complex formation. CONCLUSIONS/SIGNIFICANCE: Three main conclusions arise from our results. First, there is a genetic interaction between SEC9 and the V-ATPase, although it is unlikely that this interaction has functional significance with respect to membrane fusion or SNAREs. Second, Sro7p acts to promote SNARE complex formation. Finally, Sec9p function and SNARE complex formation are tightly coupled to the physiological state of the cell.


Subject(s)
Qc-SNARE Proteins/physiology , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Vacuolar Proton-Translocating ATPases/physiology , Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation/genetics , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , R-SNARE Proteins/genetics , R-SNARE Proteins/metabolism , SNARE Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Temperature , Vacuoles/metabolism
11.
J Thromb Haemost ; 6(10): 1757-63, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18665925

ABSTRACT

BACKGROUND: The protein SNAP-23 is part of the secretory pathway in platelets. It is, however, not entirely clear to what extent this protein contributes to the secretory function of platelets. Therefore, we overexpressed a dominant negative mutant with a novel technology that allows the creation of intact transgene-expressing platetets. RESULTS: Overexpression of a dominant negative SNAP-23 mutant that inhibited the binding of the native protein to the docking site within the secretory machinery resulted in significant suppression of the agonist-dependent surface recruitment of P-selectin and CD40L. Simultaneously, release from dense granules was clearly suppressed in the presence of this construct. Also agonist-dependent surface expression of fibrinogen receptor markers CD41 and CD61 was reduced, and agonist-triggered aggregation was inhibited. CONCLUSION: The dominant negative inhibition of SNAP-23 resulted in clear effects on platelet functions. The novel method using recombinant culture-derived platelets allowed the rapid clarification of the functional importance of this protein in intact platelets.


Subject(s)
Blood Platelets/metabolism , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Blood Platelets/physiology , CD40 Ligand , Cells, Cultured , Gene Expression , Humans , Integrin beta3 , Mutant Proteins , P-Selectin , Platelet Aggregation , Platelet Membrane Glycoprotein IIb , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , Secretory Pathway , Transfection
12.
Mol Biol Cell ; 19(9): 3769-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18579690

ABSTRACT

The assembly of four soluble N-ethylmaleimide-sensitive factor attachment protein receptor domains into a complex is essential for membrane fusion. In most cases, the four SNARE-domains are encoded by separate membrane-targeted proteins. However, in the exocytotic pathway, two SNARE-domains are present in one protein, connected by a flexible linker. The significance of this arrangement is unknown. We characterized the role of the linker in SNAP-25, a neuronal SNARE, by using overexpression techniques in synaptosomal-associated protein of 25 kDa (SNAP-25) null mouse chromaffin cells and fast electrophysiological techniques. We confirm that the palmitoylated linker-cysteines are important for membrane association. A SNAP-25 mutant without cysteines supported exocytosis, but the fusion rate was slowed down and the fusion pore duration prolonged. Using chimeric proteins between SNAP-25 and its ubiquitous homologue SNAP-23, we show that the cysteine-containing part of the linkers is interchangeable. However, a stretch of 10 hydrophobic and charged amino acids in the C-terminal half of the SNAP-25 linker is required for fast exocytosis and in its absence the calcium dependence of exocytosis is shifted toward higher concentrations. The SNAP-25 linker therefore might have evolved as an adaptation toward calcium triggering and a high rate of execution of the fusion process, those features that distinguish exocytosis from other membrane fusion pathways.


Subject(s)
Exocytosis , Gene Expression Regulation , Synaptosomal-Associated Protein 25/physiology , Amino Acid Sequence , Animals , Cattle , Chromaffin Cells/metabolism , Electrophysiology/methods , Humans , Mice , Molecular Sequence Data , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Synaptosomal-Associated Protein 25/chemistry , Synaptosomes/metabolism
13.
Biochem Biophys Res Commun ; 343(4): 1279-85, 2006 May 19.
Article in English | MEDLINE | ID: mdl-16581026

ABSTRACT

Sodium-selective amiloride-sensitive epithelial channel (ENaC) located in the apical membrane is involved in the reabsorption of sodium in tight epithelia. The soluble N-ethylmaleimide-sensitive attachment receptors (SNAREs) mediate vesicle trafficking in a variety of cell systems. Syntaxin (a t-SNARE) has been shown to interact with and functionally regulate a number of ion channels including ENaC. In this study, we investigated the role of SNAP-23, another SNARE protein, on ENaC activity in the HT-29 colonic epithelial cell system and Xenopus oocytes. Recording of amiloride-sensitive currents in both systems suggest that SNAP-23 modulates channel function, though a much higher concentration is required to inhibit ENaC in Xenopus oocytes. The introduction of Botulinum toxin A (a neurotoxin which cleaves SNAP-23), but not Botulinum toxin B or heat-inactivated Botulinum toxin A, reversed the inhibitory effect of SNAP-23 on amiloride-sensitive currents. However, syntaxin 1A and SNAP-23 combined portray a complex scenario that suggests that this channel interacts within a quaternary complex. Synaptotagmin expression neither interacts with, nor showed any effect on amiloride-sensitive currents when co-expressed with ENaC. Pull down assays suggest mild interaction between ENaC and SNAP-23, which gets stronger in the presence of syntaxin 1A. Data further suggest that SNAP-23 possibly interacts with the N-terminal alphaENaC. These functional and biochemical approaches provide evidence for a complex relationship between ENaC and the exocytotic machinery. Our data suggest that SNARE protein interplay defines the fine regulation of sodium channel function.


Subject(s)
Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/physiology , Sodium Channels/physiology , Syntaxin 1/physiology , Amiloride/pharmacology , Animals , Botulinum Toxins/pharmacology , Botulinum Toxins, Type A/pharmacology , Cell Line , Epithelial Cells , Epithelial Sodium Channels , Humans , Ion Channel Gating , Oocytes/physiology , Protein Binding , Protein Structure, Tertiary , Rats , Synaptotagmins/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Xenopus laevis
14.
J Biol Chem ; 281(48): 37091-101, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16984909

ABSTRACT

The Vam7p t-SNARE is an essential component of the vacuole fusion machinery that mediates membrane trafficking and protein sorting in yeast. Vam7p is recruited to vacuoles by its N-terminal PX domain that specifically recognizes PtdIns(3)P in the bilayers, however the precise mechanism of membrane anchoring remains unclear. Here we describe a molecular basis for membrane targeting and penetration by the Vam7p PX domain based on structural and quantitative analysis of its interactions with lipids and micelles. Our results derived from in vitro binding measurements using NMR, monolayer surface tension experiments and mutagenesis reveal a multivalent membrane docking mechanism involving specific PtdIns(3)P recognition that is facilitated by electrostatic interactions and accompanying hydrophobic insertion. Both the hydrophobic and electrostatic components enhance the Vam7p PX domain association with PtdIns(3)P-containing membranes. The inserting Val(70), Leu(71), and Trp(75) residues located next to the PtdIns(3)P binding pocket are surrounded by a basic patch, which is involved in nonspecific electrostatic contacts with acidic lipids, such as PtdSer. Substitution of the insertion residues significantly reduces the binding and penetrating power of the Vam7p PX domain and leads to cytoplasmic redistribution of the EGFP-tagged protein. The affinities of the PX domain for PtdIns(3)P and other lipids reveal a remarkable synergy within the multivalent complex that stably anchors Vam7p at the vacuolar membrane.


Subject(s)
Qc-SNARE Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Cloning, Molecular , Cytoplasm/metabolism , ErbB Receptors/metabolism , Green Fluorescent Proteins/chemistry , Leucine/chemistry , Lipids/chemistry , Magnetic Resonance Spectroscopy , Micelles , Protein Binding , Protein Structure, Tertiary , Qc-SNARE Proteins/chemistry , SNARE Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Static Electricity , Synaptosomal-Associated Protein 25 , Tryptophan/chemistry , Valine/chemistry
15.
EMBO J ; 25(22): 5260-9, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082764

ABSTRACT

Homotypic yeast vacuole fusion occurs in three stages: (i) priming reactions, which are independent of vacuole clustering, (ii) docking, in which vacuoles cluster and accumulate fusion proteins and fusion regulatory lipids at a ring-shaped microdomain surrounding the apposed membranes of each docked vacuole, where fusion will occur, and (iii) bilayer fusion/compartment mixing. These stages require vacuolar SNAREs, SNARE-chaperones, GTPases, effector complexes, and chemically minor but functionally important lipids. For each, we have developed specific ligands that block fusion and conditions that reverse each block. Using them, we test whether docking entails a linearly ordered series of catalytic events, marked by sequential acquisition of resistance to inhibitors, or whether docking subreactions are cooperative and/or reversible. We find that each fusion protein and regulatory lipid is needed throughout docking, indicative of a reversible or highly cooperative assembly of the fusion-competent vertex ring. In accord with this cooperativity, vertices enriched in one fusion catalyst are enriched in others. Docked vacuoles finally assemble SNARE complexes, yet still require physiological temperature and lipid rearrangements to complete fusion.


Subject(s)
Q-SNARE Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/physiology , Vacuoles/physiology , Catalysis , Intracellular Membranes/physiology , Ligands , Membrane Fusion , Q-SNARE Proteins/antagonists & inhibitors , Q-SNARE Proteins/genetics , Qa-SNARE Proteins/antagonists & inhibitors , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , Qb-SNARE Proteins/antagonists & inhibitors , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/physiology , Qc-SNARE Proteins/antagonists & inhibitors , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Synaptosomal-Associated Protein 25
16.
FEMS Yeast Res ; 5(11): 985-97, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16269391

ABSTRACT

We have analyzed the functions of two vacuolar t-SNAREs, Vam3p and Vam7p, in peroxisome degradation in the methylotrophic yeast Hansenula polymorpha. A Hp-vam7 mutant was strongly affected in peroxisome degradation by selective macropexophagy as well as non-selective microautophagy. Deletion of Hp-Vam3p function had only a minor effect on peroxisome degradation processes. Both proteins were located at the vacuolar membrane, with Hp-Vam7p also having a partially cytosolic location. Previously, in baker's yeast Vam3p and Vam7p have been demonstrated to be components of a t-SNARE complex essential for vacuole biogenesis. We speculate that the function of this complex in macropexophagy includes a role in membrane fusion processes between the outer membrane layer of sequestered peroxisomes and the vacuolar membrane. Our data suggest that Hp-Vam3p may be functionally redundant in peroxisome degradation. Remarkably, deletion of Hp-VAM7 also significantly affected peroxisome biogenesis and resulted in organelles with multiple, membrane-enclosed compartments. These morphological defects became first visible in cells that were in the mid-exponential growth phase of cultivation on methanol, and were correlated with accumulation of electron-dense extensions that were connected to mitochondria.


Subject(s)
Peroxisomes/metabolism , Pichia/chemistry , Vacuoles/physiology , Molecular Sequence Data , Organelles/physiology , Qa-SNARE Proteins , Qc-SNARE Proteins/physiology , SNARE Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Synaptosomal-Associated Protein 25 , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL