Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
J Cell Sci ; 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39330471

ABSTRACT

Vesicles bud from maturing Golgi cisternae in a programmed sequence. Budding is mediated by adaptors that recruit cargoes and facilitate vesicle biogenesis. In Saccharomyces cerevisiae, the AP-3 adaptor complex directs cargoes from the Golgi to the lysosomal vacuole. The AP-3 core consists of small and medium subunits complexed with two non-identical large subunits, ß3 and δ. The C termini of ß3 and δ were thought to be flexible hinges linking the core to ear domains that bind accessory proteins involved in vesicular transport. We found by computational modeling that the yeast ß3 and δ hinges are intrinsically disordered and lack folded ear domains. When either hinge is truncated, AP-3 is recruited to the Golgi, but vesicle budding is impaired, and cargoes normally sorted into the AP-3 pathway are mistargeted. This budding deficiency causes AP-3 to accumulate on ring-like Golgi structures adjacent to GGA adaptors that, in wild-type cells, bud vesicles downstream of AP-3 during Golgi maturation. Thus, each of the disordered hinges of yeast AP-3 has a crucial role in mediating transport vesicle formation at the Golgi.

2.
J Cell Biol ; 223(6)2024 06 03.
Article in English | MEDLINE | ID: mdl-38478018

ABSTRACT

The essential Golgi protein Sly1 is a member of the Sec1/mammalian Unc-18 (SM) family of SNARE chaperones. Sly1 was originally identified through remarkable gain-of-function alleles that bypass requirements for diverse vesicle tethering factors. Employing genetic analyses and chemically defined reconstitutions of ER-Golgi fusion, we discovered that a loop conserved among Sly1 family members is not only autoinhibitory but also acts as a positive effector. An amphipathic lipid packing sensor (ALPS)-like helix within the loop directly binds high-curvature membranes. Membrane binding is required for relief of Sly1 autoinhibition and also allows Sly1 to directly tether incoming vesicles to the Qa-SNARE on the target organelle. The SLY1-20 mutation bypasses requirements for diverse tethering factors but loses this ability if the tethering activity is impaired. We propose that long-range tethers, including Golgins and multisubunit tethering complexes, hand off vesicles to Sly1, which then tethers at close range to initiate trans-SNARE complex assembly and fusion in the early secretory pathway.


Subject(s)
Cytoplasmic Vesicles , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Animals , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Mammals/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Munc18 Proteins/analysis , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism , Cytoplasmic Vesicles/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism
3.
J Cell Biol ; 223(6)2024 06 03.
Article in English | MEDLINE | ID: mdl-38478017

ABSTRACT

SM proteins including Sly1 are essential cofactors of SNARE-mediated membrane fusion. Using SNARE and Sly1 mutants and chemically defined in vitro assays, we separate and assess proposed mechanisms through which Sly1 augments fusion: (i) opening the closed conformation of the Qa-SNARE Sed5; (ii) close-range tethering of vesicles to target organelles, mediated by the Sly1-specific regulatory loop; and (iii) nucleation of productive trans-SNARE complexes. We show that all three mechanisms are important and operate in parallel, and that close-range tethering promotes trans-complex assembly when cis-SNARE assembly is a competing process. Further, we demonstrate that the autoinhibitory N-terminal Habc domain of Sed5 has at least two positive activities: it is needed for correct Sed5 localization, and it directly promotes Sly1-dependent fusion. "Split Sed5," with Habc presented solely as a soluble fragment, can function both in vitro and in vivo. Habc appears to facilitate events leading to lipid mixing rather than promoting opening or stability of the fusion pore.


Subject(s)
Membrane Fusion , Munc18 Proteins , SNARE Proteins , Saccharomyces cerevisiae Proteins , Munc18 Proteins/metabolism , Protein Binding , Qa-SNARE Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism
4.
Biophys J ; 122(6): 1043-1057, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36635960

ABSTRACT

Upon nutrient limitation, budding yeast of Saccharomyces cerevisiae shift from fast growth (the log stage) to quiescence (the stationary stage). This shift is accompanied by liquid-liquid phase separation in the membrane of the vacuole, an endosomal organelle. Recent work indicates that the resulting micrometer-scale domains in vacuole membranes enable yeast to survive periods of stress. An outstanding question is which molecular changes might cause this membrane phase separation. Here, we conduct lipidomics of vacuole membranes in both the log and stationary stages. Isolation of pure vacuole membranes is challenging in the stationary stage, when lipid droplets are in close contact with vacuoles. Immuno-isolation has previously been shown to successfully purify log-stage vacuole membranes with high organelle specificity, but it was not previously possible to immuno-isolate stationary-stage vacuole membranes. Here, we develop Mam3 as a bait protein for vacuole immuno-isolation, and demonstrate low contamination by non-vacuolar membranes. We find that stationary-stage vacuole membranes contain surprisingly high fractions of phosphatidylcholine lipids (∼40%), roughly twice as much as log-stage membranes. Moreover, in the stationary stage, these lipids have higher melting temperatures, due to longer and more saturated acyl chains. Another surprise is that no significant change in sterol content is observed. These lipidomic changes, which are largely reflected on the whole-cell level, fit within the predominant view that phase separation in membranes requires at least three types of molecules to be present: lipids with high melting temperatures, lipids with low melting temperatures, and sterols.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Lipidomics , Vacuoles/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Lipids
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046036

ABSTRACT

Membranes of vacuoles, the lysosomal organelles of Saccharomyces cerevisiae (budding yeast), undergo extraordinary changes during the cell's normal growth cycle. The cycle begins with a stage of rapid cell growth. Then, as glucose becomes scarce, growth slows, and vacuole membranes phase separate into micrometer-scale domains of two liquid phases. Recent studies suggest that these domains promote yeast survival by organizing membrane proteins that play key roles in a central signaling pathway conserved among eukaryotes (TORC1). An outstanding question in the field has been whether cells regulate phase transitions in response to new physical conditions and how this occurs. Here, we measure transition temperatures and find that after an increase of roughly 15 °C, vacuole membranes appear uniform, independent of growth temperature. Moreover, populations of cells grown at a single temperature regulate this transition to occur over a surprisingly narrow temperature range. Remarkably, the transition temperature scales linearly with the growth temperature, demonstrating that the cells physiologically adapt to maintain proximity to the transition. Next, we ask how yeast adjust their membranes to achieve phase separation. We isolate vacuoles from yeast during the rapid stage of growth, when their membranes do not natively exhibit domains. Ergosterol is the major sterol in yeast. We find that domains appear when ergosterol is depleted, contradicting the prevalent assumption that increases in sterol concentration generally cause membrane phase separation in vivo, but in agreement with previous studies using artificial and cell-derived membranes.


Subject(s)
Cell Membrane/metabolism , Saccharomyces cerevisiae/physiology , Ergosterol/metabolism , Membrane Microdomains/metabolism , Temperature , Vacuoles/metabolism
6.
Traffic ; 22(1-2): 38-44, 2021 01.
Article in English | MEDLINE | ID: mdl-33225520

ABSTRACT

AP-3 (adaptor complex 3) mediates traffic from the late Golgi or early endosomes to late endosomal compartments. In mammals, mutations in AP-3 cause Hermansky-Pudlak syndrome type 2, cyclic neutropenias, and a form of epileptic encephalopathy. In budding yeast, AP-3 carries cargo directly from the trans-Golgi to the lysosomal vacuole. Despite the pathway's importance and its discovery two decades ago, rapid screens and selections for AP-3 mutants have not been available. We now report GNSI, a synthetic, genetically encoded reporter that allows rapid plate-based assessment of AP-3 functional deficiency, using either chromogenic or growth phenotype readouts. This system identifies defects in both the formation and consumption of AP-3 carrier vesicles and is adaptable to high-throughput screening or selection in both plate array and liquid batch culture formats. Episomal and integrating plasmids encoding GNSI have been submitted to the Addgene repository.


Subject(s)
Hermanski-Pudlak Syndrome , Saccharomycetales , Adaptor Protein Complex 3 , Animals , Endosomes , Transport Vesicles , Vacuoles
7.
Cell Host Microbe ; 24(2): 285-295.e8, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30057173

ABSTRACT

Many pathogenic intracellular bacteria manipulate the host phago-endosomal system to establish and maintain a permissive niche. The fate and identity of these intracellular compartments is controlled by phosphoinositide lipids. By mechanisms that have remained undefined, a Francisella pathogenicity island-encoded secretion system allows phagosomal escape and replication of bacteria within host cell cytoplasm. Here we report the discovery that a substrate of this system, outside pathogenicity island A (OpiA), represents a family of wortmannin-resistant bacterial phosphatidylinositol (PI) 3-kinase enzymes with members found in a wide range of intracellular pathogens, including Rickettsia and Legionella spp. We show that OpiA acts on the Francisella-containing phagosome and promotes bacterial escape into the cytoplasm. Furthermore, we demonstrate that the phenotypic consequences of OpiA inactivation are mitigated by endosomal maturation arrest. Our findings suggest that Francisella, and likely other intracellular bacteria, override the finely tuned dynamics of phagosomal PI(3)P in order to promote intracellular survival and pathogenesis.


Subject(s)
Francisella/growth & development , Francisella/pathogenicity , Host-Pathogen Interactions/physiology , Phagosomes/metabolism , Phagosomes/microbiology , Phosphatidylinositol 3-Kinase/metabolism , Animals , Bacterial Proteins/metabolism , Cytoplasm/microbiology , DNA Replication , Disease Models, Animal , Endosomes/microbiology , Female , Francisella/genetics , Genes, Bacterial/genetics , Genomic Islands , HEK293 Cells , HeLa Cells , Humans , Lipid Metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositols/metabolism , RAW 264.7 Cells , Type VI Secretion Systems/metabolism , Virulence Factors/metabolism
8.
Biophys J ; 113(11): 2425-2432, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29211996

ABSTRACT

Controversy has long surrounded the question of whether spontaneous lateral demixing of membranes into coexisting liquid phases can organize proteins and lipids on micron scales within unperturbed, living cells. A clear answer hinges on observation of hallmarks of a reversible phase transition. Here, by directly imaging micron-scale membrane domains of yeast vacuoles both in vivo and cell free, we demonstrate that the domains arise through a phase separation mechanism. The domains are large, have smooth boundaries, and can merge quickly, consistent with fluid phases. Moreover, the domains disappear above a distinct miscibility transition temperature (Tmix) and reappear below Tmix, over multiple heating and cooling cycles. Hence, large-scale membrane organization in living cells under physiologically relevant conditions can be controlled by tuning a single thermodynamic parameter.


Subject(s)
Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Survival , Vacuoles/metabolism , Yeasts/cytology
9.
Elife ; 62017 09 19.
Article in English | MEDLINE | ID: mdl-28925353

ABSTRACT

Zippering of SNARE complexes spanning docked membranes is essential for most intracellular fusion events. Here, we explore how SNARE regulators operate on discrete zippering states. The formation of a metastable trans-complex, catalyzed by HOPS and its SM subunit Vps33, is followed by subsequent zippering transitions that increase the probability of fusion. Operating independently of Sec18 (NSF) catalysis, Sec17 (α-SNAP) either inhibits or stimulates SNARE-mediated fusion. If HOPS or Vps33 are absent, Sec17 inhibits fusion at an early stage. Thus, Vps33/HOPS promotes productive SNARE assembly in the presence of otherwise inhibitory Sec17. Once SNAREs are partially zipped, Sec17 promotes fusion in either the presence or absence of HOPS, but with faster kinetics when HOPS is absent, suggesting that ejection of the SM is a rate-limiting step.


Subject(s)
Intracellular Membranes/physiology , Membrane Fusion , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicular Transport Proteins/metabolism
10.
Elife ; 62017 07 18.
Article in English | MEDLINE | ID: mdl-28718762

ABSTRACT

At physiological protein levels, the slow HOPS- and SNARE-dependent fusion which occurs upon complete SNARE zippering is stimulated by Sec17 and Sec18:ATP without requiring ATP hydrolysis. To stimulate, Sec17 needs its central residues which bind the 0-layer of the SNARE complex and its N-terminal apolar loop. Adding a transmembrane anchor to the N-terminus of Sec17 bypasses this requirement for apolarity of the Sec17 loop, suggesting that the loop functions for membrane binding rather than to trigger bilayer rearrangement. In contrast, when complete C-terminal SNARE zippering is prevented, fusion strictly requires Sec18 and Sec17, and the Sec17 apolar loop has functions beyond membrane anchoring. Thus Sec17 and Sec18 act twice in the fusion cycle, binding to trans-SNARE complexes to accelerate fusion, then hydrolyzing ATP to disassemble cis-SNARE complexes.


Subject(s)
Adenosine Triphosphatases/metabolism , Membrane Fusion , Proteolipids/metabolism , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adenosine Triphosphatases/chemistry , Biological Transport , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Protein Binding , Proteolipids/chemistry , SNARE Proteins/chemistry , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Vacuoles/metabolism , Vesicular Transport Proteins/chemistry
11.
Traffic ; 18(11): 720-732, 2017 11.
Article in English | MEDLINE | ID: mdl-28755404

ABSTRACT

Dense-core vesicles (DCVs) are secretory organelles that store and release modulatory neurotransmitters from neurons and endocrine cells. Recently, the conserved coiled-coil protein CCCP-1 was identified as a component of the DCV biogenesis pathway in the nematode Caenorhabditis elegans. CCCP-1 binds the small GTPase RAB-2 and colocalizes with it at the trans-Golgi. Here, we report a structure-function analysis of CCCP-1 to identify domains of the protein important for its localization, binding to RAB-2, and function in DCV biogenesis. We find that the CCCP-1 C-terminal domain (CC3) has multiple activities. CC3 is necessary and sufficient for CCCP-1 localization and for binding to RAB-2, and is required for the function of CCCP-1 in DCV biogenesis. In addition, CCCP-1 binds membranes directly through its CC3 domain, indicating that CC3 may comprise a previously uncharacterized lipid-binding motif. We conclude that CCCP-1 is a coiled-coil protein that binds an activated Rab and localizes to the Golgi via its C-terminus, properties similar to members of the golgin family of proteins. CCCP-1 also shares biophysical features with golgins; it has an elongated shape and forms oligomers.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Golgi Apparatus/metabolism , Locomotion/physiology , Secretory Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cell Membrane/metabolism , Escherichia coli/genetics , Membrane Proteins/metabolism , Neurons/metabolism , Protein Binding , Protein Domains , Protein Transport , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins
12.
Aging Cell ; 15(2): 317-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26762766

ABSTRACT

Aneuploidy and aging are correlated; however, a causal link between these two phenomena has remained elusive. Here, we show that yeast disomic for a single native yeast chromosome generally have a decreased replicative lifespan. In addition, the extent of this lifespan deficit correlates with the size of the extra chromosome. We identified a mutation in BUL1 that rescues both the lifespan deficit and a protein trafficking defect in yeast disomic for chromosome 5. Bul1 is an E4 ubiquitin ligase adaptor involved in a protein quality control pathway that targets membrane proteins for endocytosis and destruction in the lysosomal vacuole, thereby maintaining protein homeostasis. Concurrent suppression of the aging and trafficking phenotypes suggests that disrupted membrane protein homeostasis in aneuploid yeast may contribute to their accelerated aging. The data reported here demonstrate that aneuploidy can impair protein homeostasis, shorten lifespan, and may contribute to age-associated phenotypes.


Subject(s)
Aneuploidy , Saccharomyces cerevisiae/genetics , Protein Transport , Risk Factors
13.
Traffic ; 16(12): 1318-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26424513

ABSTRACT

Endosomes are transportation nodes, mediating selective transport of soluble and transmembrane cargos to and from the Golgi apparatus, plasma membrane and lysosomes. As endosomes mature to become multivesicular bodies (MVBs), Endosomal Sorting Complexes Required for Transport (ESCRTs) selectively incorporate transmembrane cargos into vesicles that bud into the endosome lumen. Luminal vesicles and their cargoes are targeted for destruction when MVBs fuse with lysosomes. Common assays of endosomal luminal targeting, including fluorescence microscopy and monitoring of proteolytic cargo maturation, possess significant limitations. We present a quantitative assay system called LUCID (LUCiferase reporter of Intraluminal Deposition) that monitors exposure of chimeric luciferase-cargo reporters to cytosol. Luciferase-chimera signal increases when sorting to the endosome lumen is disrupted, and silencing of signal from the chimera depends upon luminal delivery of the reporter rather than proteolytic degradation. The system presents several advantages, including rapidity, microscale operation and a high degree of reproducibility that enables detection of subtle phenotypic differences. Luciferase reporters provide linear signal over an extremely broad dynamic range, allowing analysis of reporter traffic even at anemic levels of expression. Furthermore, LUCID reports transport kinetics when applied to inducible trafficking reporters.


Subject(s)
Biological Assay/methods , Endosomal Sorting Complexes Required for Transport/metabolism , Lysosomes/metabolism , Multivesicular Bodies/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/metabolism , Endosomes/ultrastructure , Kinetics , Luciferases/genetics , Lysosomes/ultrastructure , Multivesicular Bodies/ultrastructure , Protein Binding , Protein Transport , Reproducibility of Results , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sensitivity and Specificity
14.
Aging (Albany NY) ; 7(6): 419-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26142908

ABSTRACT

Cellular function relies on a balance between protein synthesis and breakdown. Macromolecular breakdown through autophagy is broadly required for cellular and tissue development, function, and recovery from stress. While Caenorhabditis elegans is frequently used to explore cellular responses to development and stress, the most common assays for autophagy in this system lack tissue-level resolution. Different tissues within an organism have unique functional characteristics and likely vary in their reliance on autophagy under different conditions. To generate a tissue-specific map of autophagy in C. elegans we used a dual fluorescent protein (dFP) tag that releases monomeric fluorescent protein (mFP) upon arrival at the lysosome. Tissue-specific expression of dFP::LGG-1 revealed autophagic flux in all tissues, but mFP accumulation was most dramatic in the intestine. We also observed variable responses to stress: starvation increased autophagic mFP release in all tissues, whereas anoxia primarily increased intestinal autophagic flux. We observed autophagic flux with tagged LGG-1, LGG-2, and two autophagic cargo reporters: a soluble cytoplasmic protein, and mitochondrial TOMM-7. Finally, an increase in mFP in older worms was consistent with an age-dependent shift in proteostasis. These novel measures of autophagic flux in C. elegans reveal heterogeneity in autophagic response across tissues during stress and aging.


Subject(s)
Aging/physiology , Autophagy/physiology , Caenorhabditis elegans/physiology , Stress, Physiological/physiology , Animals , Gene Expression Regulation/physiology
15.
Proc Natl Acad Sci U S A ; 112(18): E2290-7, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902545

ABSTRACT

Sec17 [soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein; α-SNAP] and Sec18 (NSF) perform ATP-dependent disassembly of cis-SNARE complexes, liberating SNAREs for subsequent assembly of trans-complexes for fusion. A mutant of Sec17, with limited ability to stimulate Sec18, still strongly enhanced fusion when ample Sec18 was supplied, suggesting that Sec17 has additional functions. We used fusion reactions where the four SNAREs were initially separate, thus requiring no disassembly by Sec18. With proteoliposomes bearing asymmetrically disposed SNAREs, tethering and trans-SNARE pairing allowed slow fusion. Addition of Sec17 did not affect the levels of trans-SNARE complex but triggered sudden fusion of trans-SNARE paired proteoliposomes. Sec18 did not substitute for Sec17 in triggering fusion, but ADP- or ATPγS-bound Sec18 enhanced this Sec17 function. The extent of the Sec17 effect varied with the lipid headgroup and fatty acyl composition of the proteoliposomes. Two mutants further distinguished the two Sec17 functions: Sec17(L291A,L292A) did not stimulate Sec18 to disassemble cis-SNARE complex but triggered the fusion of trans-SNARE paired membranes. Sec17(F21S,M22S), with diminished apolar character to its hydrophobic loop, fully supported Sec18-mediated SNARE complex disassembly but had lost the capacity to stimulate the fusion of trans-SNARE paired membranes. To model the interactions of SNARE-bound Sec17 with membranes, we show that Sec17, but not Sec17(F21S,M22S), interacted synergistically with the soluble SNARE domains to enable their stable association with liposomes. We propose a model in which Sec17 binds to trans-SNARE complexes, oligomerizes, and inserts apolar loops into the apposed membranes, locally disturbing the lipid bilayer and thereby lowering the energy barrier for fusion.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Gene Expression Regulation, Fungal , SNARE Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicular Transport Proteins/metabolism , Adenosine Triphosphatases/chemistry , Lipid Bilayers/chemistry , Lipids/chemistry , Liposomes/chemistry , Membrane Fusion , Mutation , Protein Binding , Proteolipids/chemistry , Saccharomyces cerevisiae/metabolism
16.
Mol Biol Cell ; 26(7): 1345-56, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25673804

ABSTRACT

Vps9 and Muk1 are guanine nucleotide exchange factors (GEFs) in Saccharomyces cerevisiae that regulate membrane trafficking in the endolysosomal pathway by activating Rab5 GTPases. We show that Vps9 is the primary Rab5 GEF required for biogenesis of late endosomal multivesicular bodies (MVBs). However, only Vps9 (but not Muk1) is required for the formation of aberrant class E compartments that arise upon dysfunction of endosomal sorting complexes required for transport (ESCRTs). ESCRT dysfunction causes ubiquitinated transmembrane proteins to accumulate at endosomes, and we demonstrate that endosomal recruitment of Vps9 is promoted by its ubiquitin-binding CUE domain. Muk1 lacks ubiquitin-binding motifs, but its fusion to the Vps9 CUE domain allows Muk1 to rescue endosome morphology, cargo trafficking, and cellular stress-tolerance phenotypes that result from loss of Vps9 function. These results indicate that ubiquitin binding by the CUE domain promotes Vps9 function in endolysosomal membrane trafficking via promotion of localization.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Multivesicular Bodies/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitins/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Endosomal Sorting Complexes Required for Transport/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Saccharomyces cerevisiae Proteins/chemistry , Vesicular Transport Proteins/chemistry
18.
Elife ; 3: e02272, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24837546

ABSTRACT

Secretory and endolysosomal fusion events are driven by SNAREs and cofactors, including Sec17/α-SNAP, Sec18/NSF, and Sec1/Munc18 (SM) proteins. SMs are essential for fusion in vivo, but the basis of this requirement is enigmatic. We now report that, in addition to their established roles as fusion accelerators, SM proteins Sly1 and Vps33 directly shield SNARE complexes from Sec17- and Sec18-mediated disassembly. In vivo, wild-type Sly1 and Vps33 function are required to withstand overproduction of Sec17. In vitro, Sly1 and Vps33 impede SNARE complex disassembly by Sec18 and ATP. Unexpectedly, Sec17 directly promotes selective loading of Sly1 and Vps33 onto cognate SNARE complexes. A large thermodynamic barrier limits SM binding, implying that significant conformational rearrangements are involved. In a working model, Sec17 and SMs accelerate fusion mediated by cognate SNARE complexes and protect them from NSF-mediated disassembly, while mis-assembled or non-cognate SNARE complexes are eliminated through kinetic proofreading by Sec18.DOI: http://dx.doi.org/10.7554/eLife.02272.001.


Subject(s)
Adenosine Triphosphatases/metabolism , Munc18 Proteins/metabolism , SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Vesicular Transport Proteins/metabolism , Golgi Apparatus/metabolism , Protein Binding
19.
J Biol Chem ; 288(25): 18162-71, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23612966

ABSTRACT

VPS9 domains can act as guanosine nucleotide exchange factors (GEFs) against small G proteins of the Rab5 family. Saccharomyces cerevisiae vps9Δ mutants have trafficking defects considerably less severe than multiple deletions of the three cognate Rab5 paralogs (Vps21, Ypt52, and Ypt53). Here, we show that Muk1, which also contains a VPS9 domain, acts as a second GEF against Vps21, Ypt52, and Ypt53. Muk1 is partially redundant with Vps9 in vivo, with vps9Δ muk1Δ double mutant cells displaying hypersensitivity to temperature and ionic stress, as well as profound impairments in endocytic and Golgi endosome trafficking, including defects in sorting through the multivesicular body. Cells lacking both Vps9 and Muk1 closely phenocopy double and triple knock-out strains lacking Rab5 paralogs. Microscopy and overexpression experiments demonstrate that Vps9 and Muk1 have distinct localization determinants. These experiments establish Muk1 as the second Rab5 GEF in budding yeast.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Endocytosis , Endosomes/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins , Guanine Nucleotide Exchange Factors/genetics , Microscopy, Fluorescence , Multivesicular Bodies/metabolism , Mutation , Protein Transport/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Vacuoles/metabolism , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics
20.
Mol Biol Cell ; 23(23): 4611-22, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23051737

ABSTRACT

Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins catalyze membrane fusion events in the secretory and endolysosomal systems, and all SNARE-mediated fusion processes require cofactors of the Sec1/Munc18 (SM) family. Vps33 is an SM protein and subunit of the Vps-C complexes HOPS (homotypic fusion and protein sorting) and CORVET (class C core vacuole/endosome tethering), which are central regulators of endocytic traffic. Here we present biochemical studies of interactions between Saccharomyces cerevisiae vacuolar SNAREs and the HOPS holocomplex or Vps33 alone. HOPS binds the N-terminal H(abc) domain of the Qa-family SNARE Vam3, but Vps33 is not required for this interaction. Instead, Vps33 binds the SNARE domains of Vam3, Vam7, and Nyv1. Vps33 directly binds vacuolar quaternary SNARE complexes, and the affinity of Vps33 for SNARE complexes is greater than for individual SNAREs. Through targeted mutational analyses, we identify missense mutations of Vps33 that produce a novel set of defects, including cargo missorting and the loss of Vps33-HOPS association. Together these data suggest a working model for membrane docking: HOPS associates with N-terminal domains of Vam3 and Vam7 through Vps33-independent interactions, which are followed by binding of Vps33, the HOPS SM protein, to SNARE domains and finally to the quaternary SNARE complex. Our results also strengthen the hypothesis that SNARE complex binding is a core attribute of SM protein function.


Subject(s)
SNARE Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Vacuoles , Vesicular Transport Proteins/metabolism , Endosomes/metabolism , Endosomes/ultrastructure , Membrane Proteins/metabolism , Munc18 Proteins/metabolism , Mutation, Missense , Protein Binding , Protein Transport , Qa-SNARE Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/physiology , Synaptosomal-Associated Protein 25/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL