Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
EMBO J. 2010 Jun 16; 29(12): 1948–1960.
Published online 2010 May 14. doi: 10.1038/emboj.2010.97
PMCID: PMC2892374
PMID: 20473271

HOPS prevents the disassembly of trans-SNARE complexes by Sec17p/Sec18p during membrane fusion

Associated Data

Supplementary Materials

Abstract

SNARE-dependent membrane fusion requires the disassembly of cis-SNARE complexes (formed by SNAREs anchored to one membrane) followed by the assembly of trans-SNARE complexes (SNAREs anchored to two apposed membranes). Although SNARE complex disassembly and assembly might be thought to be opposing reactions, the proteins promoting disassembly (Sec17p/Sec18p) and assembly (the HOPS complex) work synergistically to support fusion. We now report that trans-SNARE complexes formed during vacuole fusion are largely associated with Sec17p. Using a reconstituted proteoliposome fusion system, we show that trans-SNARE complex, like cis-SNARE complex, is sensitive to Sec17p/Sec18p mediated disassembly. Strikingly, HOPS inhibits the disassembly of SNARE complexes in the trans-, but not in the cis-, configuration. This selective HOPS preservation of trans-SNARE complexes requires HOPS:SNARE recognition and is lost when the apposed bilayers are dissolved in Triton X-100; it is also observed during fusion of isolated vacuoles. HOPS thus directs the Sec17p/Sec18p chaperone system to maximize functional trans-SNARE complex for membrane fusion, a new role of tethering factors during membrane traffic.

Keywords: HOPS, membrane fusion, tethering factor, trans-SNARE complex, yeast vacuole

Introduction

Membrane fusion, the merger of two biological membranes without content leakage, is essential for protein transport along the exocytic and endocytic pathways in eukaryotic cells. Fusion requires conserved membrane-anchored proteins named SNAREs (αSNAP receptors) (Wickner and Schekman, 2008; Sudhof and Rothman, 2009), which reside on both the donor and acceptor membranes. SNAREs form four helical coiled-coil bundles through their heptad-repeat SNARE domains. Cis-SNARE complexes are disassembled by Sec17p/αSNAP and Sec18p/NSF in an ATP-dependent step called priming. Liberated SNAREs from apposed membranes form trans-SNARE complexes, an essential step for fusion. Other proteins cooperate with the SNAREs to achieve fusion. Rab GTPases and their effectors promote the tethering of donor and acceptor membranes (Grosshans et al, 2006; Markgraf et al, 2007; Hickey and Wickner, 2010) and thereby indirectly promote trans-SNARE complex formation. Sec1p/Munc18 (SM) proteins bind individual SNAREs as well as SNARE complexes and can influence fusion subreactions in either a negative or positive manner (Shen et al, 2007; Malsam et al, 2008; McNew, 2008; Deak et al, 2009).

We have studied membrane fusion using yeast vacuoles (Ungermann et al, 1998b; Collins and Wickner, 2007; Jun et al, 2007). Isolated vacuoles bear all the components required for robust fusion, including SNAREs (Vam3p, Vti1p, Vam7p, and Nyv1p), SNARE disassembly chaperones (Sec17p and Sec18p), a Rab GTPase (Ypt7p), and HOPS (Wickner, 2002). HOPS, which is composed of six subunits, fulfills multiple roles in fusion. One subunit (Vps39p) is a guanine nucleotide exchange factor for Ypt7p and another (Vps33p) belongs to the SM protein family (Seals et al, 2000; Wurmser et al, 2000). HOPS has direct affinities for Ypt7p:GTP (Seals et al, 2000), for vacuolar lipids (Stroupe et al, 2006), and for SNAREs (ibid). Each HOPS subunit is required for fusion, as mutation of its gene yields a highly fragmented vacuole morphology, indicating a fusion block (Wada et al, 1992). Antibodies to any HOPS subunit inhibit fusion (Price et al, 2000; Seals et al, 2000) and, where tested, prevent in vitro formation of trans-SNARE complexes (Price et al, 2000; Collins and Wickner, 2007).

Studies of HOPS are of general interest for understanding endomembrane traffic, because HOPS is conserved from yeast to plants to humans, and is a member of the family of large tethering factors that act at each fusion event along the secretory and endocytic pathways. In Arabidopsis thaliana, VCL1 (Vps16), AtVps11, and AtVps33 form a complex on the vacuole (Rojo et al, 2003). Mutations in Drosophila melanogaster VPS18, VPS33, or VPS41 genes alter eye colour (Pulipparacharuvil et al, 2005). In mice, the Vps33a gene regulates coat pigmentation (Suzuki et al, 2003), and human mutation in VPS33B causes arthrogryposis-renal dysfunction-cholestasis syndrome (Gissen et al, 2004). Like HOPS, which tethers endosomes and lysosomes/vacuoles, large multisubunit tethering factors regulate fusion at other organelles. CORVET acts at the endosome (Peplowska et al, 2007), the exocyst regulates cell surface growth (Guo et al, 1999), whereas COG, VFT/GARP, and TRAPP are essential for traffic to the Golgi and within that organelle (Sacher et al, 1998; Siniossoglou and Pelham, 2002; Suvorova et al, 2002). Insights about HOPS function are thus of general interest for understanding intracellular trafficking at all organelles and in all organisms.

In vacuole fusion, HOPS, along with other accessory factors such as Sec17p and Sec18p, regulate trans-SNARE complexes. In one study, HOPS suppressed the fusion of vacuoles with SNAREs bearing 0-layer alterations but not vacuoles with wild-type SNAREs (Starai et al, 2008), distinguishing cognate and noncognate SNARE complexes. Another study used vacuoles that bore lipid-anchored Nyv1p (Jun et al, 2007). These vacuoles failed to fuse, but accumulated trans-SNARE complexes that were sensitive to added Sec18p, suggesting that trans-SNARE complexes formed under these conditions are not stable. This agrees with the observations from a vacuole-mimic reconstituted proteoliposome fusion system, where vacuolar SNAREs do not suffice for trans-SNARE complex formation or fusion in the presence of Sec17p/Sec18p unless HOPS is also added (Mima et al, 2008). Using a C-terminally truncated Vam7p (Vam7p-3Δ), Schwartz and Merz (2009) captured a nonfusogenic but partially zipped trans-SNARE complex on vacuoles. Strikingly, fusion was partially restored to these vacuoles by the addition of Sec17p in a Sec18p-independent manner. A large excess of Sec18p can even inhibit vacuole fusion (Ungermann et al, 1998b). Taken together, these studies show that Sec17p, Sec18p, and HOPS have access to trans-SNARE complexes.

We now report that the trans-SNARE complex formed during vacuole fusion is largely associated with Sec17p. Using reconstituted proteoliposomes, we demonstrate that trans-SNARE complex is subject to disassembly by Sec17p/Sec18p. HOPS inhibits the disassembly of the trans-SNARE complex, but not the cis-SNARE complex, and the integrity of the apposed membranes is required for this aspect of HOPS function. Our findings explain the synergy between Sec17p/Sec18p and HOPS for membrane fusion (Mima et al, 2008); Sec17p/Sec18p disassemble cis-SNARE complexes, but productive trans-SNARE complexes are protected by HOPS. As all membrane fusion events involve Sec17p/αSNAP, Sec18p/NSF, SNAREs, large tethering complexes, and SM proteins, a similar mechanism may regulate trans-SNARE complexes on other organelles.

Results

Sec17p is associated with Vam3p during vacuole fusion

To determine the partners of the trans-SNARE complex, we searched for proteins that bind Vam3p in a fusion pathway dependent manner. Vacuoles from BJ3505 nyv1Δ (which has full-length Vam3p) and DKY6281 vam3(ΔN) (bearing N-terminally deleted Vam3p) were isolated and used in fusion reactions (Figure 1A). Neither initial vacuole population has Nyv1p associated with full-length Vam3p. After co-incubation, we prepared detergent extracts and immunoprecipitated the full-length Vam3p by using antibodies that are specific for the N-terminal domain of Vam3p. Sypro Ruby staining revealed several proteins that were specifically associated with Vam3p under fusion conditions (Figure 1B and C, lane 1). Mass spectrometry analysis (http://yeastrc.org/pdr/) indicated that these proteins include Nyv1p, Vti1p, Vam7p, and Sec17p.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f1.jpg

Sec17p associates with Vam3p during fusion. (A) Assay scheme. Fusion reactions containing BJ3505 nyv1Δ vacuoles, DKY6281 vam3(ΔN) vacuoles, and 40 nM Vam7p were incubated at 27°C for 45 min with either no inhibitors, Gyp1-46 (117 μg/ml)/Gdi1p (30 μg/ml), or MED (10 μM). At the end of the reaction, Pho8p activity was assayed (B). As control, BJ and DKY vacuoles were incubated separately, and the Pho8p activity from each was measured and added (lane 4). The rest of the vacuoles were solubilized in detergent and proteins co-immunoprecipitated with αVam3 were subject to SDS–PAGE and Sypro Ruby staining (C) and to mass spectrometry analysis. Asterisks in (C) indicate unique bands. The arrow points to the Sec17p band. (D) The Sec17p association with Vam3p (without additional Vam7p) was assayed. BJ3505 nyv1Δ vacuoles (33 μg) and DKY6281 vam3(ΔN) vacuoles (33 μg) were incubated for 45 min with indicated reagents (117 μg/ml Gyp1-46, 30 μg/ml Gdi1p, 64 μg/ml αVps33, 75 μg/ml αYpt7, 67 μg/ml Ypt7 peptide) and 6 μg of vacuoles were analysed for Pho8p activity. The remainder was solubilized in detergent (see Materials and methods). Nyv1p and Sec17p that co-immunoprecipitated with Vam3p were analysed by immunoblot. Two regions of the same blot from one gel are shown; there is an empty space between lanes 5 and 6. See Supplementary Figure S1 for the original scan.

Sec17p on vacuoles interacts with individual SNAREs as well as with the SNARE complex (Collins et al, 2005). However, Sec17p is released from vacuoles in the presence of Sec18p and ATP, an early event in the vacuole fusion reaction (Mayer et al, 1996), and Sec17p is not recovered with Vam3p when fusion (but not priming) is blocked (Figure 1D, lanes 6, 7, and 9). A portion of the Sec17p appeared to be recruited to Vam3p under conditions that permit fusion and the formation of new SNARE complexes (Figure 1D, lanes 5 and 8).

Trans-SNARE complex is largely associated with Sec17p

To determine whether Sec17p was associating with free Vam3p or with Vam3p in a SNARE complex, we again used vacuoles from BJ3505 nyv1Δ and DKY6281 vam3(ΔN) strains. At the end of the fusion reaction, we immunodepleted Sec17p from the vacuolar detergent extracts and asked whether SNARE complexes, as assayed by Nyv1p that was bound to full-length Vam3p, were removed through their association with Sec17p (Figure 2A). When fusion is robust (Figure 2B, lane I), virtually all the newly formed SNARE complexes are removed by Sec17p immunodepletion (Figure 2C, left panel, compare lane 10 with lane 9 or 11), showing that most of the newly formed SNARE complexes interact with Sec17p.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f2.jpg

Trans-SNARE complex is largely associated with Sec17p. (A) Assay scheme. Vacuoles from BJ3505 nyv1Δ (93 μg) were incubated with vacuoles (93 μg) from DKY6281 vam3(ΔN) or DKY6281 vam3(ΔN) nyv1-CCIIM under standard fusion conditions for 45 min before an aliquot was assayed for Pho8p activity (B). The rest were solubilized in detergent and divided equally into three portions, each to be incubated with agarose resin conjugated with either αPorin I (a control IgG), αSec17p, or no IgG. After 1 h incubation, supernatant was collected by centrifugation and incubated with fresh resin for the second time to ensure complete depletion of Sec17p (lanes 3, 7, 14, and 18). A small aliquot of the supernatant collected after the second incubation was set aside for western blotting, whereas the rest were incubated with immobilized αVam3-N. Proteins co-precipitated with Vam3p were subject to SDS–PAGE and immunoblot analysis (C). Note that there are more Nyv1 molecules on DKY6281 vam3(ΔN) vacuoles than on DKY6281 vam3(ΔN) nyv1-CCIIM vacuoles (compare lanes 1 and 12), as reported earlier (Jun et al, 2007), but the amount of new SNARE complex formed in reactions I and II are comparable (compare lanes 11 and 22).

At least three types of SNARE complex form during these incubations: trans-SNARE complex, cis-SNARE complex that was trans- before fusion, and cis-SNARE complex that formed de novo after fusion and had never been trans-. To determine whether Sec17p is associated with the trans-SNARE complex, we exploited vacuoles from DKY6281 vam3(ΔN) nyv1-CCIIM. The transmembrane domain of Nyv1p on these vacuoles had been replaced by the Ykt6p prenylation motif (Jun et al, 2007). Vacuoles with lipid-anchored Nyv1p fuse poorly (Figure 2B, lane II) and thus a larger percentage of the new SNARE complex will be trans-SNARE complex as compared with reactions involving vacuoles with the wild-type Nyv1p. Earlier studies using Nyv1p-CCIIM vacuoles (Figure 6E of Jun et al, 2007) established that >50% of the new SNARE complex formed is trans-SNARE complex. Remarkably, immunoadsorption of Sec17p completely depleted the new SNARE complex from detergent extracts of docked NYV1-CCIIM vacuoles (Figure 2B, compare lane 21 with lane 20 or 22), showing that the new SNARE complex, including the trans-SNARE complex, is largely associated with Sec17p. What is the significance of the interaction between Sec17p and the trans-SNARE complex?

Trans-SNARE complex is sensitive to Sec17p/Sec18p

Earlier studies have shown that a partially lipid-anchored trans-SNARE complex formed during vacuole fusion is subject to Sec18p and Vam7p-dependent remodelling, implicating Sec17p (along with Sec18p) in disassembling the trans-SNARE complex (Jun et al, 2007). To study SNAREs with wild-type membrane anchors, we have exploited a reconstituted proteoliposome fusion system made from recombinant proteins and a set of pure lipids that closely match the composition of vacuole membranes (Mima et al, 2008). Fusion is measured by lipid mixing between proteoliposomes (Figure 3A and B). By placing Nyv1p on one set of liposomes and Vam3p and Vti1p on the other, we are able to bypass Sec17p/Sec18p-dependent priming and directly study new SNARE complex formation. In this assay, fusion (Figure 3A) and new SNARE complex formation (Figure 3C) require the addition of the soluble SNARE Vam7p. Both fusion and SNARE complex formation are stimulated by HOPS. It is noteworthy that similar rates of fusion can be achieved despite drastically different amounts of SNARE complex (compare Figure 3C, lanes 6 and 10, and the corresponding fusion in Figure 3A, filled circles and filled inverted triangles, respectively), suggesting that not all new SNARE complexes are functionally equal.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f3.jpg

A truncated Vam7p (Vam7p-3Δ) arrests the fusion of proteoliposomes while allowing trans-SNARE complex formation. Proteoliposomes bearing Nyv1p were mixed with proteoliposomes carrying Vam3p and Vti1p. Wild-type Vam7p, Vam7p-3Δ, and/or HOPS (61 nM) was added as indicated (A, B). Fusion between donor and acceptor liposomes was measured by the dequenching of NBD-PE fluorescence (see Materials and methods). (C) At the end of the reaction, proteoliposomes were solubilized in detergent, and the proteins that adsorbed to immobilized αVam3 were analysed by SDS–PAGE and immunoblot.

To resolve trans-SNARE complex formation from fusion and possible formation of post-fusion cis-SNARE complexes, we used a C-terminally truncated Vam7p (Vam7p-3Δ) that efficiently drives trans-SNARE complex formation on isolated vacuoles in the absence of fusion (Schwartz and Merz, 2009). In the reconstituted fusion system, Vam7p-3Δ supports trans-SNARE complex formation (Figure 3C, lanes 12–17) without fusion (Figure 3B). The yield of trans-SNARE complex increases with increasing amounts of Vam7p-3Δ in the reaction. In addition, HOPS promotes the formation of trans-SNARE complexes in the presence of Vam7p-3Δ (Figure 3C). Vam7p-3Δ therefore permits study of trans-SNARE complex without the interference of post-fusion cis-SNARE complexes.

To determine whether trans-SNARE complexes can be disassembled by Sec17p and Sec18p, reactions were divided into two stages, assembly of trans-SNARE complexes followed by their disassembly by Sec17p/Sec18p. During the first stage incubation, 6% of the Nyv1p entered into trans-SNARE complex with Vam3p in the presence of Vam7p-3Δ (Figure 4, compare lanes 5 and 13). If the reaction was allowed to continue for another 30 min (second stage), more trans-SNARE complex was detected (Figure 4, compare lane 7 and 13). Sec17p and Sec18p added at the beginning of the second stage reduced the amount of Nyv1p associated with Vam3p to about 2% (Figure 4, lane 9), significantly below the level reached in the first stage (6%), showing that Sec17p and Sec18p disassemble the complex already formed in the first stage. Please note that under our immunoprecipitation conditions, the Sec17p/Sec18p is inactivated by EDTA/RIPA buffer (Supplementary Figure S2), so that any SNARE complex disassembly we measure has occurred on the membrane rather than in detergent.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f4.jpg

Trans-SNARE complex is disassembled by Sec17p/Sec18p. Donor proteoliposomes (50 μM) bearing Nyv1p were mixed with acceptor proteoliposomes (400 μM) bearing Vam3p and Vti1p (with 1 mM MgCl2, 1 mM ATP and an ATP regenerating system). Samples received either 1.2 μM Vam7p-3Δ, 1.2 μM Vam7p-3Δ with 12 μM FYVE, or control buffer. After 30 min at 27°C (Stage I), samples with Vam7p-3Δ were either transferred to ice or incubated at 27°C for another 30 min without Sec17p/Sec18p (lanes 7 and 10) or with the addition of Sec17p/Sec18p (0.34 μM/0.24 μM or 0.68 μM/0.24 μM; lanes 8, 9, 11, and 12). Samples from lanes 10 to 12 also received FYVE at the end of Stage I to prevent the formation of new SNARE complex. At the end of Stage II, Vam7p-3Δ (1.2 μM) was added to the sample as shown in lane 1, which had not initially received Vam7p-3Δ. All samples were assayed for trans-SNARE complex (see Materials and methods). The Nyv1p signal, normalized for the recovery of Vam3p, was averaged from three independent experiments. Error bars represent standard error of the mean (s.e.m.) calculated using KaleidaGraph. Note that lane 13 measures trans-SNARE complex formation in the first stage only. If the second stage reaction continues at 27°C without inhibitors, more trans-SNARE complex will form (compare lanes 13 and 7).

To control for SNARE complex assembly during the second stage of the reaction, we also performed this stage of the reaction with purified recombinant FYVE domain. FYVE domain inhibits vacuole fusion by binding to PI(3)P (Fratti et al, 2004), a regulatory lipid that is essential for trans-SNARE complex formation (Collins and Wickner, 2007). As FYVE domain inhibits the trans-SNARE complex formation on proteoliposomes (Figure 4, lane 6 versus 7), adding FYVE domain at the start of the second stage of the reaction allows direct evaluation of the effect of Sec17p and Sec18p on existing complexes. As shown in Figure 4 (lanes 10–12), Sec17p/Sec18p disassembles trans-SNARE complex in a concentration-dependent manner. This explains why Sec17p and Sec18p block the fusion of proteoliposomes in the absence of HOPS (Mima et al, 2008).

HOPS inhibits the Sec17/18-dependent disassembly of the trans-SNARE complex

To allow SNARE-dependent membrane fusion in the presence of Sec17p and Sec18p, the disassembly of trans-SNARE complexes must be inhibited. HOPS and Sec17p/Sec18p synergistically promote proteoliposome fusion (Mima et al, 2008), suggesting that HOPS might regulate the Sec17p/Sec18p-mediated disassembly of trans-SNARE complex. As mentioned above, we allowed trans-SNARE complex to form in an initial incubation in the presence or absence of HOPS. To arrest further trans-SNARE complex assembly while examining HOPS function, we could not use FYVE domain as in Figure 4, as FYVE binds to phosphoinositides that are part of the HOPS membrane receptor (Burd and Emr, 1998; Kutateladze et al, 1999). We therefore added either CaCl2 (10 mM) or KCl (250 mM), which effectively prevents fusion (data not shown) and trans-SNARE complex formation (Supplementary Figures S3 and S4). Without HOPS, trans-SNARE complexes are disassembled by Sec17p and Sec18p in the second stage of the reaction (lanes 9–12 in Figure 5A and B). The disassembly rate is slower in the presence of 250 mM KCl, presumably due to salt inhibition of Sec17p/Sec18p. Strikingly, trans-SNARE complexes are quite resistant to Sec17p/Sec18p-mediated disassembly in the presence of HOPS (lanes 5–8 in Figure 5A and B). There is a slight decrease of trans-SNARE complex at high concentrations of Sec17p/Sec18p (compare lane 8 with lane 5), suggesting that the HOPS inhibition of trans-SNARE complex disassembly is not absolute.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f5.jpg

HOPS inhibits the disassembly of trans-SNARE complex by Sec17p/Sec18p. Donor proteoliposomes (50 μM) bearing Nyv1p, acceptor proteoliposomes (400 μM) bearing Vam3p and Vti1p, and 1.2 μM Vam7p-3Δ were mixed with 61 nM HOPS or its buffer alone. Trans-SNARE complex was allowed to form at 27°C for 30 min. Samples then received either 10 mM CaCl2 (A) or 250 mM KCl (B) to stop the formation of new SNARE complex. Samples were then divided into four portions and received either Sec17p/Sec18p (0.68 μM/0.24 μM or 1.36 μM/0.48 μM) (lanes 7, 8, 11, and 12) or control buffer (lanes 5, 6, 9, and 10). After 30 min of further incubation at 27°C (except samples in lanes 6 and 10, which were placed on ice), trans-SNARE complex was assayed. Data from three experiments were analysed as in Figure 4. In both (A, B), the P-values between lanes 5 and 8 or 5 and 7 are larger than 0.25.

Sec17p and Sec18p can disassemble a complex of four vacuolar SNARE soluble domains and this disassembly is not affected by HOPS (Mima et al, 2008), suggesting that membrane integrity might be required for the HOPS-dependent preservation of the trans-SNARE complex. To directly address this, we modified the trans-SNARE complex disassembly assay by also preparing a parallel set of samples, which received Triton X-100 during the second stage of the reaction (Figure 6A, lanes 7–12). Triton X-100 solubilization of donor and acceptor membranes allowed Vam3p (from acceptor proteoliposome) and Nyv1p (from donor proteoliposomes) that had not been in trans-SNARE complex to form a complex in detergent despite the addition of excess soluble GST-Nyv1p to compete with Nyv1p. This led to the detection of a modest increase of Vam3p–Nyv1p association (compare Figure 6A, lanes 1 and 7, lanes 4 and 10). Strikingly, almost all these complexes were effectively disassembled by Sec17p/Sec18p regardless of the presence of HOPS (compare Figure 6A, lanes 7 and 9, lanes 10 and 12). Trans-SNARE complexes formed on apposed liposomes that were not subsequently exposed to detergent maintained their resistance to Sec17p/Sec18p in the presence of HOPS (Figure 6A, lanes 1–6). Thus, the HOPS-dependent preservation of trans-SNARE complexes appears to rely on the integrity of the two apposed membranes.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f6.jpg

HOPS preservation of trans-SNARE complex requires membrane integrity (A) and HOPS:Vam7p interaction (B). (A) trans-SNARE complex was formed at 27°C for 30 min as described in Figure 5. After receiving 10 mM CaCl2, aliquots received various amounts of Sec17p/Sec18p (0/0, 0.68 μM/0.24 μM, or 1.36 μM/0.48 μM) with (lanes 7–12) or without (lanes 1–6) 1% Triton X-100. After 30 min of further incubation at 27°C, trans-SNARE complex was assayed. Data from five independent experiments were analysed. Error bars represent s.e.m. For samples in lanes 7–12, GST-Nyv1p was added before the addition of Triton X-100 to reduce new SNARE complex formation in detergent. (B) Proteoliposomes bearing Nyv1p (50 μM) and proteoliposomes (400 μM) bearing Vam3p and Vti1p were mixed with 1.2 μM Vam7p-3Δ at 27°C for 30 min to allow trans-SNARE complex formation. After receiving 10 mM CaCl2, samples were aliquoted into tubes containing HOPS, HOPS with Y42A-PX, or control buffers. The final concentration of Y42A-PX is 10.8 μM, and thus the molar ratio of Y42A-PX:Vam7p-3Δ=9:1. Each aliquot was further divided into three portions to receive Sec17p/Sec18p (0/0, 0.68 μM/0.24 μM, or 1.36 μM/0.48 μM). After 30 min of additional incubation at 27°C, trans-SNARE complex was assayed. Data from four independent experiments were analysed. Error bars represent s.e.m.

To address whether SNARE recognition is required for this new function of HOPS, we examined the effect of saturating HOPS for its capacity to bind SNAREs. We have previously shown that HOPS interacts preferentially with Vam7p among the vacuolar SNAREs, and that this interaction is specific for the N-terminal PX domain of Vam7p (Stroupe et al, 2006). We therefore tested whether the addition of an excess of this PX domain would block HOPS preservation of trans-SNARE complexes. To avoid complications arising from the affinity of this PX domain for vacuolar PI(3)P, we used the Y42A mutant PX domain, which specifically lacks affinity for phosphoinositides (Cheever et al, 2001). Proteoliposomes bearing Vam3p and Vti1p were incubated with those bearing Nyv1p in the presence of Vam7p-3Δ to allow trans-SNARE complex assembly, then mixed with CaCl2 to prevent further trans-SNARE complex assembly. After adding combinations of HOPS and/or Y42A-PX, a second incubation was performed that included Sec17p and Sec18p where indicated. The trans-SNARE complexes formed during the initial incubation were readily disassembled by added Sec17p/Sec18p (Figure 6B, lanes 4–6), and HOPS inhibited this disassembly (Figure 6B, lanes 1–3). Adding Y42A-PX alone had no effect on the Sec17p/Sec18p-mediated disassembly (Figure 6B, lanes 10–12), but it prevented HOPS from preserving the trans-SNARE complex (Figure 6B, lanes 7–9). Thus, HOPS appears to rely on both membrane bilayer integrity (Figure 6A) and on its interaction with SNAREs (Figure 6B) to preserve trans-SNARE complexes.

HOPS does not inhibit cis-SNARE complex disassembly

Is this effect of HOPS specific for trans-SNARE complexes or does HOPS also inhibit cis-SNARE complex disassembly by Sec17p/Sec18p? Proteoliposomes bearing Vam3p, Vam7p, Vti1p, and Nyv1p were incubated at low concentration to eliminate tethering and trans-SNARE complex formation (see Materials and methods). The disassembly of their cis-SNARE complexes was measured by the loss of association between Nyv1p and Vam3p. Approximately 60% of the Nyv1p on these proteoliposomes was initially associated with Vam3p (Figure 7A). Incubation with Sec17p and Sec18p reduced the level of Nyv1p associated with Vam3p to 25%. The complexes that were insensitive to Sec17p and Sec18p were likely oriented toward the liposome lumen and thus inaccessible to the SNARE chaperones. HOPS did not inhibit cis-SNARE complex disassembly at any Sec17p/Sec18p concentration tested (Figure 7A).

An external file that holds a picture, illustration, etc.
Object name is emboj201097f7.jpg

HOPS does not inhibit cis-SNARE complex disassembly. (A) 4-SNARE proteoliposomes (100 μM lipids), 61 nM HOPS or its buffer, and various amounts of Sec17p/Se18p (0/0, 0.17 μM/0.06 μM, 0.34 μM/0.12 μM, or 0.68 μM/0.24 μM) were incubated at 27°C for 30 min. The Nyv1p bound to Vam3p was assayed as described (see Materials and methods). Data from three independent experiments were analysed as in Figure 4. (B, C) To control for the effect of HOPS pre-incubation with proteoliposomes and to use the same amount SNARE complex as in the trans-SNARE complex disassembly assay, reduced amounts of 4-SNARE proteoliposomes (3.3 μM lipids) bearing either Vam7p (B) or Vam7p-3Δ (C) were incubated with 61 nM HOPS or its buffer alone at 27°C for 30 min. Aliquots received control buffer or various amounts of Sec17p/Sec18p as in Figure 7A. After 30 min of further incubation at 27°C, cis-SNARE complex was assayed. The amount of cis-SNARE complex detected in the absence of Sec17p/Sec18p was set at 100% and was used to normalize the cis-SNARE complexes detected in the presence of Sec17p/Sec18p. Data from four independent experiments were analysed. Error bars represent s.e.m. At each Sec17p/Sec18p concentration, the differences between samples with or without HOPS were not statistically significant.

To more closely mimic the conditions used in the trans-SNARE complex disassembly assays (Figure 5) and, in particular, to account for the fact that the proportion of the SNAREs engaged in trans complexes (4–8% of total; Figures 5 and and6)6) is smaller than that of the cis-SNARE complex (60% Figure 7A) in our reactions, we lowered the amount of 4-SNARE proteoliposomes in the cis-SNARE disassembly assay so that the concentrations of the SNARE complexes in the two were equivalent (see Material and methods). 4-SNARE proteoliposomes that bear either Vam7p-3Δ or Vam7p were pre-incubated with HOPS (or control buffer) for 30 min before the addition of Sec17p and Sec18p at the same concentrations used in the trans-SNARE complex disassembly assay. Sec17p and Sec18p effectively disassembled cis-SNARE complexes in the presence or absence of HOPS (Figure 7B and C). Thus, the HOPS-mediated preservation of trans-SNARE complexes, which requires the integrity of the two apposed membranes, is specific to the trans conformation of the SNARE complex.

HOPS relieves Sec18-dependent inhibition of vacuole fusion

Is the HOPS-dependent preservation of trans-SNARE complexes on proteoliposomes bearing only pure fusion proteins also seen on the intact and chemically complex organelle? The fusion of vacuoles can be modulated by exogenous Sec18p (Figure 8A), as reported (Ungermann et al, 1998b). At low concentrations, Sec18p stimulates fusion, suggesting that vacuolar cis-SNARE complexes are not completely disassembled by the endogenous Sec18p. However, high Sec18p concentrations are inhibitory. As most vacuolar trans-SNARE complex is associated with Sec17p (Figure 2), the excess Sec18p may rapidly disassemble trans-SNARE complexes before they can promote membrane fusion. In accord with this idea, the addition of HOPS relieved the inhibitory effect of excessive Sec18p (Figure 8B). Vacuoles normally bear sufficient HOPS for fusion, and thus standard vacuole fusion reactions were not affected by the further addition of 10–40 nM HOPS. However, at even 10 nM, added HOPS prevented the inhibition by Sec18p. The fusion signal from reactions containing both Sec18p and HOPS is slightly higher, probably because Sec18p disassembled cis-SNARE complexes to promote fusion while not interfering with trans-SNARE complex function. Thus, HOPS preserves trans-SNARE complexes on the intact organelle as well as with reconstituted proteoliposomes.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f8.jpg

HOPS prevents Sec18p-mediated inhibition of vacuole fusion. (A) Fusion reactions containing BJ3505 and DKY6281 vacuoles were incubated at 27°C for 60 min with increasing amounts of Sec18p and compensatory amounts of Sec18p buffer. At the end of the reaction, Pho8p activity was assayed. (B) BJ3505 and DKY6281 vacuoles were mixed with either 0.57 μM Sec18p or control buffer, along with various amount of HOPS and compensatory amounts of HOPS buffer. Pho8p activity was assayed after 60 min incubation at 27°C (except for the ice control), Pho8p activity was assayed. Fusion signals, after subtracting the ice background signal, were averaged from three independent experiments. Error bars represent s.e.m.

Discussion

Although HOPS is required for vacuole fusion, its mechanism of synergy with Sec17p/Sec18p has been unclear. Sec17p/Sec18p can disassemble either cis-SNARE complexes (Figure 7; Ungermann et al, 1998a) or trans-SNARE complexes (Figures 4, ,55 and and6;6; Jun et al, 2007), and HOPS will associate with either cis-SNARE complexes (Collins et al, 2005) or trans-SNARE complexes (Figure 6B). HOPS promotes the tethering of vacuoles (Stroupe et al, 2006) or reconstituted proteoliposomes (Stroupe et al, 2009). Tethering is a prerequisite for trans-SNARE complex formation (Hickey and Wickner, 2010). It is not known whether HOPS directly catalyses trans-SNARE complex formation or makes the trans-SNARE complex more potent for fusion. We now report that HOPS inhibits the disassembly of trans-SNARE complexes by Sec17p and Sec18p, placing HOPS near the SNAREs at a step immediately before fusion. Trans-SNARE complexes may undergo multiple cycles of assembly and disassembly to allow the accumulation of cognate, functional trans-SNARE complexes. HOPS could create a physical barrier to stabilize functional trans-SNARE complexes against attack by Sec17p/Sec18p, allowing fusion to occur (Figure 9). HOPS association with trans-SNARE complex may be important for this protection. As both trans- and cis-SNARE complexes presumably have the same cytoplasmic helical bundle, it is reasonable that the selective protection of trans-SNARE complex by HOPS depends on the intact, apposed membranes of tethered vesicles (Figure 6). We hypothesize that HOPS protects the trans-SNARE complex only when it binds to both SNAREs and to the apposed membranes (Figure 9). Such a topology does not exist for cis-SNARE complexes, which are formed by SNAREs on the same membrane, suggesting why cis-SNARE complexes are not protected by HOPS. As HOPS can proofread several structural features of trans-SNARE complexes, it may only protect functional trans-SNARE complexes.

An external file that holds a picture, illustration, etc.
Object name is emboj201097f9.jpg

Model of HOPS action. SNAREs on apposed biological membranes can form a trans-SNARE complex that is accessible to Sec17p and Sec18p in the absence of HOPS. On ATP hydrolysis, Sec17p and Sec18p can disassemble the trans-SNARE complex and thus prevent the merger of the two membranes. HOPS, by binding simultaneously to two apposed membranes, protects trans-SNARE complexes from Sec17p and Sec18p. The block is relieved on fusion as HOPS no longer binds two membranes, allowing post-fusion cis-SNARE complex to be primed for a second round of fusion. This model does not exclude other HOPS functions, such as tethering and proofreading.

A second, alternative model to explain HOPS action is that its association with trans-SNARE complexes causes a conformational change that renders the SNAREs resistant to Sec17p/Sec18p mediated disassembly, and this conformational change would reverse if the HOPS:trans-SNARE complex were solubilized in Triton X-100. We note, however, that Triton X-100 preserves the structure and activity of many membrane complexes, and specifically allows HOPS to remain with SNARE complexes (Collins et al, 2005). Further studies will be required to test these two models.

The dynamic nature of the vacuolar trans-SNARE complex is not limited to proteoliposomes. Trans-SNARE complexes on vacuoles can undergo Sec17p and Sec18p-dependent remodelling during fusion (Jun et al, 2007). The finding that trans-SNARE complexes formed during vacuole fusion are largely associated with Sec17p (Figure 1) and that excessive Sec18p inhibits vacuole fusion (Figure 8; Ungermann et al, 1998b) suggests that most of these complexes are subject to remodelling. However, trans-SNARE complex remodelling is not essential for fusion under all circumstances. Fusion persists after Sec17p/Sec18p are inactivated or removed (Mayer et al, 1996), and both vacuoles and proteoliposomes are capable of fusion without Sec17p and Sec18p at all (Figure 3A; Thorngren et al, 2004; Mima et al, 2008). Thus, trans-SNARE complex remodelling is not absolutely required for merger of two biological membranes. However, wild-type vacuoles, which bear noncognate SNAREs such as Snc2p (Robinson et al, 2006; Collins and Wickner, 2007), are always subject to Sec17p and Sec18p action in vivo. Rapid trans-SNARE complex turnover coupled with selective stabilization by HOPS may optimize vacuole fusion in the cell.

Trans-SNARE complex remodelling may not be unique to vacuole fusion. Exocytosis in neuroendocrine PC-12 cells is blocked by additional αSNAP after priming and docking, and extra NSF can relieve the blockade (Barszczewski et al, 2008), suggesting potential roles for these SNARE chaperones at a stage well beyond cis-SNARE complex disassembly. Studies of sea urchin eggs indicated that SNARE complexes formed between purified secretory vesicles can be disassembled before fusion (Tahara et al, 1998), though purposeful cycles of disassembly and reassembly have not been reported in this system. Reconstituted proteoliposome fusion systems offer advantages for mechanistic studies. In addition to vacuolar SNAREs, neuronal (Weber et al, 1998) and endosomal (Ohya et al, 2009) SNAREs have been successfully reconstituted into proteoliposomes. The neuronal trans-SNARE complex is functionally resistant to αSNAP and NSF (Weber et al, 2000). When a self-inhibitory domain of syntaxin was removed, fusion was accelerated by the addition of αSNAP and NSF (Weber et al, 2000). This is in contrast to proteoliposomes bearing vacuolar SNAREs, which do not fuse at all in the presence of Sec17p and Sec18p without HOPS (Mima et al, 2008). The difference may be attributable to the different lipid composition in the two proteoliposome fusion systems or to differences in the neuronal and vacuolar SNARE complexes, for example, neuronal trans-SNARE complexes may execute fusion at a rate faster than the rate of turnover by αSNAP and NSF. Reconstituted early endosome fusion requires a set of 17 recombinant proteins including SNAREs, αSNAP and NSF, Rab5 and their partners (Ohya et al, 2009). The first round of fusion takes place without αSNAP and NSF, because VAMP4 and sytaxin13/VTI1A/syntaxin6 are reconstituted separately on two different proteoliposomes. When added individually, αSNAP or NSF blocked this fusion, presumably either by preventing SNAREs from entering into trans-SNARE complexes or by inhibiting trans-SNARE complex function. A trans-SNARE complex assay similar to the one used in our study might distinguish these possibilities. Like reconstituted vacuolar fusion, the αSNAP/NSF-dependent endosomal fusion requires a protein complex that contains a Vps33p isoform, human VPS45. Biochemical analysis will be needed to address whether this complex preserves the endosomal trans-SNARE complex as HOPS does for the vacuole.

Recent studies of intra-Golgi traffic have shown that the COG tethering complex, which is required for the stability of the intro-Golgi SNARE complex (Shestakova et al, 2007), must interact with Sly1p, a Vps33 isoform, for Golgi SNARE-pairing (Laufman et al, 2009). If COG, like HOPS, is indeed preserving the trans-SNARE complex, then this new activity may be a common feature of oligomeric tethering factors.

Materials and methods

Yeast strains, vacuole isolation, and fusion reactions

Vacuoles were purified (Haas et al, 1994) from Saccharomyces cerevisiae strains BJ3505, DKY6281, BJ3505 nyv1Δ, DKY6281 vam3(ΔN), and DKY6281 vam3(ΔN) nyv1-CCIIM (Jun et al, 2007) for fusion and trans-SNARE complex assays. Fusion reactions (30 μl) contained 20 mM PIPES-KOH, pH 6.8, 200 mM sorbitol, 125 mM KCl, 6 mM MgCl2, 1 mM ATP, an ATP regenerating system (1 mg/ml creatine kinase and 29 mM creatine phosphate), 10 mM coenzyme A, 3.3 μg/ml Pbi2p (I2B), 3 μg of pep4Δ vacuoles (from BJ3505 derivatives), and 3 μg of pho8Δ vacuoles (from DKY6281 derivatives). After incubation at 27°C, Pho8p phosphatase activity was assayed at 30°C for 5 min (Haas, 1995). Fusion units are micromoles of p-nitrophenol phosphate hydrolyzed per min per microgram pep4Δ vacuole.

Proteins, antibodies, and cross-linking

Vam7p and Vam7p-3Δ were purified according to Schwartz and Merz (2009) with the following modifications: (1) Escherichia coli Rosetta 2 (DE3) (Novagen) was used for protein expression; (2) induced bacteria were lysed by French Press (3 passes, 900 pounds per square inch); and (3) after affinity purification on Ni-NTA agarose (Qiagen), his6-Vam7 proteins were exchanged into RB500 (20 mM HEPES-NaOH, pH 7.4, 500 mM NaCl, 10% (v/v) glycerol) for storage. Recombinant Vam3p, Vti1p, and Nyv1p were purified from Rosetta 2 (Novagen) (Mima et al, 2008). HOPS complex containing Vps33-GST was purified from yeast CHY31 (Starai et al, 2008). Sec17p was purified from Rosetta 2 (DE3) as described (Schwartz and Merz, 2009), except that the tagless Sec17p was exchanged into RB150 (20 mM HEPES-NaOH, pH 7.4, 150 mM NaCl, 10% (v/v) glycerol) for storage. His6-Sec18 (Haas and Wickner, 1996), I2B (Slusarewicz et al, 1997), GST-2xFYVE domain (Gillooly et al, 2000), his6-Gyp1-46 (Wang et al, 2003), Gdi1p (Starai et al, 2007), Y42A-PX domain (Boeddinghaus et al, 2002), αYpt7, and its antigenic peptides (Eitzen et al, 2001), αVPS33 (Seals et al, 2000), and αSec17 (Haas and Wickner, 1996) were prepared as described. αPorin I was a kind gift from Naomi Thorngren. MARCKS effector domain (Wang et al, 2001) was synthesized by the WM Keck Biotechnology Resource Center (New Haven, CT).

αVam3 and αVam3-N (specific for N-terminal domain) were affinity purified with immobilized GST-Vam3(ΔTM) and GST-Vam3-N(ΔTM), respectively. Proteins (10 mg) were cross-linked to 2.5 ml of Sulfolink coupling gel (Thermo Scientific, Rockford, IL) following the manufacturer's instructions. Serum (10 ml) from rabbits immunized with his6-Vam3(ΔTM) was used for antibody purification (Jun et al, 2007).

For immunoprecipitation assays, antibodies (αVam3, αVam3-N, αSec17, or αPorin I) were cross-linked to protein A Sepharose CL-4B (GE) as described (Harlow and Lane, 1998), except that 2 mg of IgG (1 OD280=0.8 mg/ml) were used per 4 ml of protein A resin. Protein A resin cross-linked to antibodies was stored at 4°C in PBS containing 0.02% Na3N. Immediately before use, resins were exchanged into elution buffer (0.2 M glycine, pH 2.2, 0.2 M NaCl), then equilibrated with solubilization buffer (see below).

Proteoliposome reconstitution and lipid mixing assay

SNARE proteoliposomes with POPC (42 or 44% for donor or acceptor), POPE (18%), soy PI (18%), POPS (4.4%), POPA (2.0%), CL (1.6%), ERG (8.0%), DAG (1.0%), PI(3)P (1.0%), PI(4,5)P2 (1.0%), and fluorescent lipids (1.5% NBD-PE and 1.5% Rh-PE for donor; 1.0% dansyl-PE for acceptor) were prepared as described (Mima et al, 2008).

Fusion reactions (20 μl) contained RB150 (20 mM HEPES-NaOH, pH 7.4, 150 mM NaCl, 10% (v/v) glycerol), 1 mM MgCl2, 1 mM ATP, an ATP regenerating system, donor proteoliposomes (50 μM lipids), and acceptor proteoliposomes (400 μM lipids). Vam7p or Vam7-3Δ (0.2–1.8 μM), HOPS (61 nM), Sec17p (0.17–2.7 μM), and Sec18p (0.06–0.96 μM) were added where indicated. To monitor lipid mixing, reaction mixtures (prepared on ice) were transferred to a 396-well plate (pre-incubated on ice) and the NBD fluorescent signal was measured (λex=460 nm, λem=538 nm, λex=515 nm) in a SpectraMAX Geminin XPS plate reader (Molecular Devices) at 27°C for 30 min (Mima et al, 2008). Fusion is presented as the ratio of the fluorescence at any time to that at 1 min.

Trans-SNARE complex assay

Proteoliposome samples (450 μM lipids; 20 μl) were chilled (ice, 5 min). Each received 1.4 μg of GST-Nyv1(ΔTM) and, after an additional 10 min, 800 μl of RIPA buffer (25 mM Tris–Cl, pH 7.5, 150 mM NaCl, 1% NP40-Alternative, 1% deoxycholate, 0.1% SDS) containing 10 mM EDTA, protease inhibitor cocktail (0.46 μg/ml leupeptin, 3.5 μg/ml pepstatin A, 24 μg/ml pefablock-SC), and 1 mM PMSF. GST-Nyv1(ΔTM) (70 μg/ml) was added to block potential binding of native Nyv1p to Vam3p in detergent. Samples were nutated at 4°C for 20 min, then centrifuged (16 000 g, 4°C, 5 min). Supernatant (700 μl) was incubated with 15 μl of immobilized αVam3 at 4°C for 1 h with nutation. After centrifugation (3000 g, 4°C, 2 min), the αVam3 resin was resuspended in 600 μl of RIPA buffer (with 10 mM EDTA and protease inhibitor cocktail), centrifuged (3000 g, 4°C, 2 min), and the supernatant was removed by centrifugation. After repeating this process three times, proteins were eluted with 2 × sample buffer (Sambrook et al, 1989) and subjected to SDS–PAGE (12%) and western blot analysis. The blots were scanned and analysed using software UN-SCAN-IT gel 5.3 (Silk Scientific).

To assay for vacuolar trans-SNARE complex, vacuoles from a 300-μl reaction were centrifuged (11 750 g, 10 min, 4°C) and resuspended in 400 μl of solubilization buffer (20 mM Tris–Cl, pH 7.5, 150 mM NaCl, 1 mM MgCl2, 10% glycerol, 0.5% NP40-Alternative) containing protease inhibitor cocktail and 1 mM PMSF. After 20 min at 4°C, samples were centrifuged (16 000 g, 20 min, 4°C) and supernatant (350 μl) was incubated with 30 μl αVam3-N resin at 4°C overnight. Resin was resuspended in 600 μl of solubilization buffer with protease inhibitor cocktail, centrifuged (3000 g, 4°C, 2 min), and the supernatant was removed. This process was repeated three times before proteins were eluted from the resin and assayed as above. For immunoadsorption experiments, vacuolar detergent extracts were incubated with resin bearing either αSec17 or αPorin I (30 μl) at 4°C for 1 h. Supernatant was collected after centrifugation (3000 g, 4°C, 2 min) and incubated (4°C, 1 h) with fresh resin and again centrifuged. Supernatant was then used for overnight incubation with αVam3-N resin as described above. For experiments in Figure 1, a larger fusion reaction (4.83 ml) containing BJ3505 nyv1Δ vacuoles (483 μg), DKY6281 vam3(ΔN) vacuoles (483 μg), and 40 nM Vam7p was incubated at 27°C for 45 min with either no inhibitors, Gyp1-46 (2.5 μM)/Gdi1p (0.6 μM) or MED (10 μM). Aliquots (30 μl) were set aside for Pho8p activity assay. The remainder was resuspended in 6.4 ml solubilization buffer (described above). Detergent extracts were incubated with 480 μl of immobilized αVam3(N) at 4°C overnight. Proteins were eluted with 6 ml of 0.2 M glycine (pH 2.2), 0.2 M NaCl and divided into two equal aliquots. Ice-cold TCA (100%) was added to each to 12.5%. After vigorous mixing (5 s), samples were incubated on ice for 30 min and centrifuged (16 000 g, 4°C, 20 min). The pellet in each aliquot was resuspended in 3 ml of acetone (−20°C) and centrifuged (16 000 g, 4°C, 10 min). Supernatants were removed and pellets were dried in air for 5 min. One aliquot was subjected to SDS–PAGE and Sypro Ruby staining and the other to mass spectrometry.

Multidimensional protein identification technology

The protein digest was pressure loaded onto a fused silica capillary desalting column containing 5 cm of 5 μm Polaris C18-A material (Metachem, Ventura, CA) packed into a 250-μm internal diameter (i.d.) capillary with a 2-μm filtered union (UpChurch Scientific, Oak Harbor, WA). The desalting column was washed with buffer containing 95% water, 5% acetonitrile, and 0.1% formic acid. After desalting, a 100-μm i.d. capillary with a 5-μm pulled tip packed with 10 cm 3-μm Aqua C18 material (Phenomenex, Ventura, CA) followed by 3 cm 5-μm Partisphere strong cation exchanger (Whatman, Clifton, NJ) was attached to the filter union and the entire split column (desalting column–filter union–analytical column) was placed inline with an Agilent 1100 quaternary HPLC (Palo Alto, CA) and analysed using a modified five-step separation described earlier (Washburn et al, 2001). The buffer solutions used were 5% acetonitrile/0.1% formic acid (buffer A), 80% acetonitrile/0.1% formic acid (buffer B), and 500 mM ammonium acetate/5% acetonitrile/0.1% formic acid (buffer C). Step 1 consisted of a 90-min gradient from 0 to 100% buffer B. Steps 2–4 had the following profile: 3 min of 100% buffer A, 2 min of X% buffer C, a 10-min gradient from 0 to 15% buffer B, and a 97-min gradient from 15 to 45% buffer B. The 2-min buffer C percentages (X) were 20, 50, 80%, respectively, for the five-step analysis. The final step, the gradient contained: 3 min of 100% buffer A, 20 min of 100% buffer C, a 10-min gradient from 0 to 15% buffer B, and a 107-min gradient from 15 to 70% buffer B.

As peptides eluted from the microcapillary column, they were electrosprayed directly into an LTQ two-dimensional ion trap mass spectrometer (ThermoFinnigan, Palo Alto, CA) with the application of a distal 2.4 kV spray voltage. A cycle of one full-scan mass spectrum (400–1400 m/z) followed by eight data-dependent MS/MS spectra at a 35% normalized collision energy was repeated continuously throughout each step of the multidimensional separation. Application of mass spectrometer scan functions and HPLC solvent gradients were controlled by the Xcalibur data system.

Analysis of tandem mass spectra

MS/MS spectra were analysed using the following software analysis protocol. Poor quality spectra were removed from the data set using an automated spectral quality assessment algorithm (Bern et al, 2004). MS/MS spectra remaining after filtering were searched with the SEQUESTTM algorithm (Eng et al, 1994) against the Saccharomyces Genome Database (December 2005) concatenated to a decoy database in which the sequence for each entry in the original database was reversed (Peng et al, 2003). All searches were parallelized and performed on a Beowulf computer cluster consisting of 100 1.2 GHz Athlon CPUs (Sadygov et al, 2002). No enzyme specificity was considered for any search. SEQUEST results were assembled and filtered using the DTASelect (version 2.0) program (Tabb et al, 2002; Cociorva et al, 2007). DTASelect 2.0 uses a linear discriminant analysis to dynamically set XCorr and DeltaCN thresholds for the entire data set to achieve a user-specified false-positive rate (5% in this analysis). The false-positive rates are estimated by the program from the number and quality of spectral matches to the decoy database. All the data sets can be found under VAM3 in the Yeast Resource Center Public Data Repository (Riffle et al, 2005).

Cis-SNARE complex disassembly assay

Proteoliposomes (100 or 3.3 μM lipids) bearing Vam3p, Vti1p, Vam7p (either wild-type or truncated), and Nyv1p were incubated in 20 μl with RB150, 1 mM MgCl2, 1 mM ATP, an ATP regenerating system, and (where indicated) HOPS, Sec17p, and Sec18p. After 30 min at 27°C, samples were chilled on ice for 5 min before receiving 2.8 μg of GST-Nyv1p (ΔTM) and 400 μl of RIPA buffer containing 10 mM EDTA, protease inhibitor cocktail, and 1 mM PMSF. After 20 min at 4°C, samples were centrifuged (16 000 g, 5 min, 4°C), and the supernatant (350 μl) was collected for immunoprecipitation as described above.

Supplementary Material

Supplementary Information:
Review Process File:

Acknowledgments

We thank Alex Merz (U Washington) for sharing Vam7p and Vam7p-3Δ constructs before publication and Amy Orr and Holly Jakubowski for excellent technical support. This work was supported by NIH grant GM23377. Mass spectrometry analysis was supported by NIH grant P41 RR11823. Hao Xu and William Wickner designed the experiments and wrote the manuscript. Youngsoo Jun conceived the trans-SNARE complex assay and engineered the required yeast strains. James Thompson and John Yates performed mass spectrometry analysis and wrote the corresponding methods. William Wickner isolated SNARE proteins and the lyticase used for vacuole isolation. Hao Xu performed the rest of the experiments.

Footnotes

The authors declare that they have no conflict of interest.

References

  • Barszczewski M, Chua JJ, Stein A, Winter U, Heintzmann R, Zilly FE, Fasshauer D, Lang T, Jahn R (2008) A novel site of action for alpha-SNAP in the SNARE conformational cycle controlling membrane fusion. Mol Biol Cell 19: 776–784 [PMC free article] [PubMed] [Google Scholar]
  • Bern M, Goldberg D, McDonald WH, Yates JR III (2004) Automatic quality assessment of peptide tandem mass spectra. Bioinformatics 20 (Suppl 1): i49–i54 [PubMed] [Google Scholar]
  • Boeddinghaus C, Merz AJ, Laage R, Ungermann C (2002) A cycle of Vam7p release from and PtdIns 3-P-dependent rebinding to the yeast vacuole is required for homotypic vacuole fusion. J Cell Biol 157: 79–89 [PMC free article] [PubMed] [Google Scholar]
  • Burd CG, Emr SD (1998) Phosphatidylinositol(3)-phosphate signaling mediated by specific binding to RING FYVE domains. Mol Cell 2: 157–162 [PubMed] [Google Scholar]
  • Cheever ML, Sato TK, de Beer T, Kutateladze TG, Emr SD, Overduin M (2001) Phox domain interaction with PtdIns(3)P targets the Vam7 t-SNARE to vacuole membranes. Nat Cell Biol 3: 613–618 [PubMed] [Google Scholar]
  • Cociorva D, L Tabb D, Yates JR (2007) Validation of tandem mass spectrometry database search results using DTASelect. Curr Protoc Bioinformatics Chapter 13: Unit 13.4 [PubMed] [Google Scholar]
  • Collins KM, Thorngren NL, Fratti RA, Wickner WT (2005) Sec17p and HOPS, in distinct SNARE complexes, mediate SNARE complex disruption or assembly for fusion. EMBO J 24: 1775–1786 [PMC free article] [PubMed] [Google Scholar]
  • Collins KM, Wickner WT (2007) Trans-SNARE complex assembly and yeast vacuole membrane fusion. Proc Natl Acad Sci USA 104: 8755–8760 [PMC free article] [PubMed] [Google Scholar]
  • Deak F, Xu Y, Chang WP, Dulubova I, Khvotchev M, Liu X, Sudhof TC, Rizo J (2009) Munc18-1 binding to the neuronal SNARE complex controls synaptic vesicle priming. J Cell Biol 184: 751–764 [PMC free article] [PubMed] [Google Scholar]
  • Eitzen G, Thorngren N, Wickner W (2001) Rho1p and Cdc42p act after Ypt7p to regulate vacuole docking. EMBO J 20: 5650–5656 [PMC free article] [PubMed] [Google Scholar]
  • Eng J, McCormack A, Yates J (1994) An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J Am Soc Mass Spectrom 5: 976–989 [PubMed] [Google Scholar]
  • Fratti RA, Jun Y, Merz AJ, Margolis N, Wickner W (2004) Interdependent assembly of specific regulatory lipids and membrane fusion proteins into the vertex ring domain of docked vacuoles. J Cell Biol 167: 1087–1098 [PMC free article] [PubMed] [Google Scholar]
  • Gillooly DJ, Morrow IC, Lindsay M, Gould R, Bryant NJ, Gaullier JM, Parton RG, Stenmark H (2000) Localization of phosphatidylinositol 3-phosphate in yeast and mammalian cells. EMBO J 19: 4577–4588 [PMC free article] [PubMed] [Google Scholar]
  • Gissen P, Johnson CA, Morgan NV, Stapelbroek JM, Forshew T, Cooper WN, McKiernan PJ, Klomp LW, Morris AA, Wraith JE, McClean P, Lynch SA, Thompson RJ, Lo B, Quarrell OW, Di Rocco M, Trembath RC, Mandel H, Wali S, Karet FE et al. (2004) Mutations in VPS33B, encoding a regulator of SNARE-dependent membrane fusion, cause arthrogryposis-renal dysfunction-cholestasis (ARC) syndrome. Nat Genet 36: 400–404 [PubMed] [Google Scholar]
  • Grosshans BL, Ortiz D, Novick P (2006) Rabs and their effectors: achieving specificity in membrane traffic. Proc Natl Acad Sci USA 103: 11821–11827 [PMC free article] [PubMed] [Google Scholar]
  • Guo W, Roth D, Walch-Solimena C, Novick P (1999) The exocyst is an effector for Sec4p, targeting secretory vesicles to sites of exocytosis. EMBO J 18: 1071–1080 [PMC free article] [PubMed] [Google Scholar]
  • Haas A (1995) A quantitative assay to measure homotypic vacuole fusion in vitro. Methods Cell Sci 17: 283–294 [Google Scholar]
  • Haas A, Conradt B, Wickner W (1994) G-protein ligands inhibit in vitro reactions of vacuole inheritance. J Cell Biol 126: 87–97 [PMC free article] [PubMed] [Google Scholar]
  • Haas A, Wickner W (1996) Homotypic vacuole fusion requires Sec17p (yeast alpha-SNAP) and Sec18p (yeast NSF). EMBO J 15: 3296–3305 [PMC free article] [PubMed] [Google Scholar]
  • Harlow E, Lane D (1998) Using Antibodies: A Laboratory Manual. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press [Google Scholar]
  • Hickey CM, Wickner W (2010) HOPS initiates vacuole docking by tethering membranes prior to trans-SNARE complex assembly. Mol Biol Cell (in press) [PMC free article] [PubMed] [Google Scholar]
  • Jun Y, Xu H, Thorngren N, Wickner W (2007) Sec18p and Vam7p remodel trans-SNARE complexes to permit a lipid-anchored R-SNARE to support yeast vacuole fusion. EMBO J 26: 4935–4945 [PMC free article] [PubMed] [Google Scholar]
  • Kutateladze TG, Ogburn KD, Watson WT, de Beer T, Emr SD, Burd CG, Overduin M (1999) Phosphatidylinositol 3-phosphate recognition by the FYVE domain. Mol Cell 3: 805–811 [PubMed] [Google Scholar]
  • Laufman O, Kedan A, Hong W, Lev S (2009) Direct interaction between the COG complex and the SM protein, Sly1, is required for Golgi SNARE pairing. EMBO J 28: 2006–2017 [PMC free article] [PubMed] [Google Scholar]
  • Malsam J, Kreye S, Sollner TH (2008) Membrane fusion: SNAREs and regulation. Cell Mol Life Sci 65: 2814–2832 [PMC free article] [PubMed] [Google Scholar]
  • Markgraf DF, Peplowska K, Ungermann C (2007) Rab cascades and tethering factors in the endomembrane system. FEBS Lett 581: 2125–2130 [PubMed] [Google Scholar]
  • Mayer A, Wickner W, Haas A (1996) Sec18p (NSF)-driven release of Sec17p (alpha-SNAP) can precede docking and fusion of yeast vacuoles. Cell 85: 83–94 [PubMed] [Google Scholar]
  • McNew JA (2008) Regulation of SNARE-mediated membrane fusion during exocytosis. Chem Rev 108: 1669–1686 [PubMed] [Google Scholar]
  • Mima J, Hickey CM, Xu H, Jun Y, Wickner W (2008) Reconstituted membrane fusion requires regulatory lipids, SNAREs and synergistic SNARE chaperones. EMBO J 27: 2031–2042 [PMC free article] [PubMed] [Google Scholar]
  • Ohya T, Miaczynska M, Coskun U, Lommer B, Runge A, Drechsel D, Kalaidzidis Y, Zerial M (2009) Reconstitution of Rab- and SNARE-dependent membrane fusion by synthetic endosomes. Nature 459: 1091–1097 [PubMed] [Google Scholar]
  • Peng J, Elias JE, Thoreen CC, Licklider LJ, Gygi SP (2003) Evaluation of multidimensional chromatography coupled with tandem mass spectrometry (LC/LC-MS/MS) for large-scale protein analysis: the yeast proteome. J Proteome Res 2: 43–50 [PubMed] [Google Scholar]
  • Peplowska K, Markgraf DF, Ostrowicz CW, Bange G, Ungermann C (2007) The CORVET tethering complex interacts with the yeast Rab5 homolog Vps21 and is involved in endo-lysosomal biogenesis. Dev Cell 12: 739–750 [PubMed] [Google Scholar]
  • Price A, Wickner W, Ungermann C (2000) Proteins needed for vesicle budding from the Golgi complex are also required for the docking step of homotypic vacuole fusion. J Cell Biol 148: 1223–1229 [PMC free article] [PubMed] [Google Scholar]
  • Pulipparacharuvil S, Akbar MA, Ray S, Sevrioukov EA, Haberman AS, Rohrer J, Kramer H (2005) Drosophila Vps16A is required for trafficking to lysosomes and biogenesis of pigment granules. J Cell Sci 118 (Part 16): 3663–3673 [PubMed] [Google Scholar]
  • Riffle M, Malmstrom L, Davis TN (2005) The yeast resource center public data repository. Nucleic Acids Res 33 (Database issue): D378–D382 [PMC free article] [PubMed] [Google Scholar]
  • Robinson M, Poon PP, Schindler C, Murray LE, Kama R, Gabriely G, Singer RA, Spang A, Johnston GC, Gerst JE (2006) The Gcs1 Arf-GAP mediates Snc1,2 v-SNARE retrieval to the Golgi in yeast. Mol Biol Cell 17: 1845–1858 [PMC free article] [PubMed] [Google Scholar]
  • Rojo E, Zouhar J, Kovaleva V, Hong S, Raikhel NV (2003) The AtC-VPS protein complex is localized to the tonoplast and the prevacuolar compartment in arabidopsis. Mol Biol Cell 14: 361–369 [PMC free article] [PubMed] [Google Scholar]
  • Sacher M, Jiang Y, Barrowman J, Scarpa A, Burston J, Zhang L, Schieltz D, Yates JR III, Abeliovich H, Ferro-Novick S (1998) TRAPP, a highly conserved novel complex on the cis-Golgi that mediates vesicle docking and fusion. EMBO J 17: 2494–2503 [PMC free article] [PubMed] [Google Scholar]
  • Sadygov RG, Eng J, Durr E, Saraf A, McDonald H, MacCoss MJ, Yates JR III (2002) Code developments to improve the efficiency of automated MS/MS spectra interpretation. J Proteome Res 1: 211–215 [PubMed] [Google Scholar]
  • Sambrook J, Fritsch E, Maniatis T (1989) Molecular Cloning: A Laboratory Manual. Cold Spring Harbor, NY: Cold Spring Harbor Press [Google Scholar]
  • Schwartz ML, Merz AJ (2009) Capture and release of partially zipped trans-SNARE complexes on intact organelles. J Cell Biol 185: 535–549 [PMC free article] [PubMed] [Google Scholar]
  • Seals DF, Eitzen G, Margolis N, Wickner WT, Price A (2000) A Ypt/Rab effector complex containing the Sec1 homolog Vps33p is required for homotypic vacuole fusion. Proc Natl Acad Sci USA 97: 9402–9407 [PMC free article] [PubMed] [Google Scholar]
  • Shen J, Tareste DC, Paumet F, Rothman JE, Melia TJ (2007) Selective activation of cognate SNAREpins by Sec1/Munc18 proteins. Cell 128: 183–195 [PubMed] [Google Scholar]
  • Shestakova A, Suvorova E, Pavliv O, Khaidakova G, Lupashin V (2007) Interaction of the conserved oligomeric Golgi complex with t-SNARE Syntaxin5a/Sed5 enhances intra-Golgi SNARE complex stability. J Cell Biol 179: 1179–1192 [PMC free article] [PubMed] [Google Scholar]
  • Siniossoglou S, Pelham HR (2002) Vps51p links the VFT complex to the SNARE Tlg1p. J Biol Chem 277: 48318–48324 [PubMed] [Google Scholar]
  • Slusarewicz P, Xu Z, Seefeld K, Haas A, Wickner WT (1997) I2B is a small cytosolic protein that participates in vacuole fusion. Proc Natl Acad Sci USA 94: 5582–5587 [PMC free article] [PubMed] [Google Scholar]
  • Starai VJ, Hickey CM, Wickner W (2008) HOPS proofreads the trans-SNARE complex for yeast vacuole fusion. Mol Biol Cell 19: 2500–2508 [PMC free article] [PubMed] [Google Scholar]
  • Starai VJ, Jun Y, Wickner W (2007) Excess vacuolar SNAREs drive lysis and Rab bypass fusion. Proc Natl Acad Sci USA 104: 13551–13558 [PMC free article] [PubMed] [Google Scholar]
  • Stroupe C, Collins KM, Fratti RA, Wickner W (2006) Purification of active HOPS complex reveals its affinities for phosphoinositides and the SNARE Vam7p. EMBO J 25: 1579–1589 [PMC free article] [PubMed] [Google Scholar]
  • Stroupe C, Hickey CM, Mima J, Burfeind AS, Wickner W (2009) Minimal membrane docking requirements revealed by reconstitution of Rab GTPase-dependent membrane fusion from purified components. Proc Natl Acad Sci USA 106: 17626–17633 [PMC free article] [PubMed] [Google Scholar]
  • Sudhof TC, Rothman JE (2009) Membrane fusion: grappling with SNARE and SM proteins. Science 323: 474–477 [PMC free article] [PubMed] [Google Scholar]
  • Suvorova ES, Duden R, Lupashin VV (2002) The Sec34/Sec35p complex, a Ypt1p effector required for retrograde intra-Golgi trafficking, interacts with Golgi SNAREs and COPI vesicle coat proteins. J Cell Biol 157: 631–643 [PMC free article] [PubMed] [Google Scholar]
  • Suzuki T, Oiso N, Gautam R, Novak EK, Panthier JJ, Suprabha PG, Vida T, Swank RT, Spritz RA (2003) The mouse organellar biogenesis mutant buff results from a mutation in Vps33a, a homologue of yeast vps33 and Drosophila carnation. Proc Natl Acad Sci USA 100: 1146–1150 [PMC free article] [PubMed] [Google Scholar]
  • Tabb DL, McDonald WH, Yates JR III (2002) DTASelect and Contrast: tools for assembling and comparing protein identifications from shotgun proteomics. J Proteome Res 1: 21–26 [PMC free article] [PubMed] [Google Scholar]
  • Tahara M, Coorssen JR, Timmers K, Blank PS, Whalley T, Scheller R, Zimmerberg J (1998) Calcium can disrupt the SNARE protein complex on sea urchin egg secretory vesicles without irreversibly blocking fusion. J Biol Chem 273: 33667–33673 [PubMed] [Google Scholar]
  • Thorngren N, Collins KM, Fratti RA, Wickner W, Merz AJ (2004) A soluble SNARE drives rapid docking, bypassing ATP and Sec17/18p for vacuole fusion. EMBO J 23: 2765–2776 [PMC free article] [PubMed] [Google Scholar]
  • Ungermann C, Nichols BJ, Pelham HR, Wickner W (1998a) A vacuolar v-t-SNARE complex, the predominant form in vivo and on isolated vacuoles, is disassembled and activated for docking and fusion. J Cell Biol 140: 61–69 [PMC free article] [PubMed] [Google Scholar]
  • Ungermann C, Sato K, Wickner W (1998b) Defining the functions of trans-SNARE pairs. Nature 396: 543–548 [PubMed] [Google Scholar]
  • Wada Y, Ohsumi Y, Anraku Y (1992) Genes for directing vacuolar morphogenesis in Saccharomyces cerevisiae. I. Isolation and characterization of two classes of vam mutants. J Biol Chem 267: 18665–18670 [PubMed] [Google Scholar]
  • Wang J, Arbuzova A, Hangyas-Mihalyne G, McLaughlin S (2001) The effector domain of myristoylated alanine-rich C kinase substrate binds strongly to phosphatidylinositol 4,5-bisphosphate. J Biol Chem 276: 5012–5019 [PubMed] [Google Scholar]
  • Wang L, Merz AJ, Collins KM, Wickner W (2003) Hierarchy of protein assembly at the vertex ring domain for yeast vacuole docking and fusion. J Cell Biol 160: 365–374 [PMC free article] [PubMed] [Google Scholar]
  • Washburn MP, Wolters D, Yates JR III (2001) Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat Biotechnol 19: 242–247 [PubMed] [Google Scholar]
  • Weber T, Parlati F, McNew JA, Johnston RJ, Westermann B, Sollner TH, Rothman JE (2000) SNAREpins are functionally resistant to disruption by NSF and alphaSNAP. J Cell Biol 149: 1063–1072 [PMC free article] [PubMed] [Google Scholar]
  • Weber T, Zemelman BV, McNew JA, Westermann B, Gmachl M, Parlati F, Sollner TH, Rothman JE (1998) SNAREpins: minimal machinery for membrane fusion. Cell 92: 759–772 [PubMed] [Google Scholar]
  • Wickner W (2002) Yeast vacuoles and membrane fusion pathways. EMBO J 21: 1241–1247 [PMC free article] [PubMed] [Google Scholar]
  • Wickner W, Schekman R (2008) Membrane fusion. Nat Struct Mol Biol 15: 658–664 [PMC free article] [PubMed] [Google Scholar]
  • Wurmser AE, Sato TK, Emr SD (2000) New component of the vacuolar class C-Vps complex couples nucleotide exchange on the Ypt7 GTPase to SNARE-dependent docking and fusion. J Cell Biol 151: 551–562 [PMC free article] [PubMed] [Google Scholar]

Articles from The EMBO Journal are provided here courtesy of Nature Publishing Group

-