Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
Mol Biol Cell ; 35(5): ar63, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38446621

ABSTRACT

Regulation of the luminal pH of late endocytic compartments in continuously fed mammalian cells is poorly understood. Using normal rat kidney fibroblasts, we investigated the reversible assembly/disassembly of the proton pumping V-ATPase when endolysosomes are formed by kissing and fusion of late endosomes with lysosomes and during the subsequent reformation of lysosomes. We took advantage of previous work showing that sucrosomes formed by the uptake of sucrose are swollen endolysosomes from which lysosomes are reformed after uptake of invertase. Using confocal microscopy and subcellular fractionation of NRK cells stably expressing fluorescently tagged proteins, we found net recruitment of the V1 subcomplex during sucrosome formation and loss during lysosome reformation, with a similar time course to RAB7a loss. Addition of invertase did not alter mTORC1 signalling, suggesting that the regulation of reversible V-ATPase assembly/disassembly in continuously fed cells differs from that in cells subject to amino acid depletion/refeeding. Using live cell microscopy, we demonstrated recruitment of a fluorescently tagged V1 subunit during endolysosome formation and a dynamic equilibrium and rapid exchange between the cytosolic and membrane bound pools of this subunit. We conclude that reversible V-ATPase assembly/disassembly plays a key role in regulating endolysosomal/lysosomal pH in continuously fed cells.


Subject(s)
Vacuolar Proton-Translocating ATPases , Rats , Animals , Vacuolar Proton-Translocating ATPases/metabolism , beta-Fructofuranosidase/metabolism , Endosomes/metabolism , Signal Transduction , Lysosomes/metabolism , Mammals/metabolism
2.
Hum Mutat ; 43(12): 2265-2278, 2022 12.
Article in English | MEDLINE | ID: mdl-36153662

ABSTRACT

A rare and fatal disease resembling mucopolysaccharidosis in infants, is caused by impaired intracellular endocytic trafficking due to deficiency of core components of the intracellular membrane-tethering protein complexes, HOPS, and CORVET. Whole exome sequencing identified a novel VPS33A mutation in a patient suffering from a variant form of mucopolysaccharidosis. Electron and confocal microscopy, immunoblotting, and glycosphingolipid trafficking experiments were undertaken to investigate the effects of the mutant VPS33A in patient-derived skin fibroblasts. We describe an attenuated juvenile form of VPS33A-related syndrome-mucopolysaccharidosis plus in a man who is homozygous for a hitherto unknown missense mutation (NM_022916.4: c.599 G>C; NP_075067.2:p. Arg200Pro) in a conserved region of the VPS33A gene. Urinary glycosaminoglycan (GAG) analysis revealed increased heparan, dermatan sulphates, and hyaluronic acid. We showed decreased abundance of VPS33A in patient derived fibroblasts and provided evidence that the p.Arg200Pro mutation leads to destablization of the protein and proteasomal degradation. As in the infantile form of mucopolysaccharidosis plus, the endocytic compartment in the fibroblasts also expanded-a phenomenon accompanied by increased endolysosomal acidification and impaired intracellular glycosphingolipid trafficking. Experimental treatment of the patient's cultured fibroblasts with the proteasome inhibitor, bortezomib, or exposure to an inhibitor of glucosylceramide synthesis, eliglustat, improved glycosphingolipid trafficking. To our knowledge this is the first report of an attenuated juvenile form of VPS33A insufficiency characterized by appreciable residual endosomal-lysosomal trafficking and a milder mucopolysaccharidosis plus than the disease in infants. Our findings expand the proof of concept of redeploying clinically approved drugs for therapeutic exploitation in patients with juvenile as well as infantile forms of mucopolysaccharidosis plus disease.


Subject(s)
Mutation, Missense , Vesicular Transport Proteins , Humans , Male , Endosomes/metabolism , Lysosomes/metabolism , Mutation , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
3.
Traffic ; 23(5): 238-269, 2022 05.
Article in English | MEDLINE | ID: mdl-35343629

ABSTRACT

Since the discovery of lysosomes more than 70 years ago, much has been learned about the functions of these organelles. Lysosomes were regarded as exclusively degradative organelles, but more recent research has shown that they play essential roles in several other cellular functions, such as nutrient sensing, intracellular signalling and metabolism. Methodological advances played a key part in generating our current knowledge about the biology of this multifaceted organelle. In this review, we cover current methods used to analyze lysosome morphology, positioning, motility and function. We highlight the principles behind these methods, the methodological strategies and their advantages and limitations. To extract accurate information and avoid misinterpretations, we discuss the best strategies to identify lysosomes and assess their characteristics and functions. With this review, we aim to stimulate an increase in the quantity and quality of research on lysosomes and further ground-breaking discoveries on an organelle that continues to surprise and excite cell biologists.


Subject(s)
Lysosomes , Metabolic Networks and Pathways , Lysosomes/metabolism , Signal Transduction
4.
J Cell Sci ; 134(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34042162

ABSTRACT

To provide insights into the kiss-and-run and full fusion events resulting in endocytic delivery to lysosomes, we investigated conditions causing increased tethering and pore formation between late endocytic organelles in HeLa cells. Knockout of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) VAMP7 and VAMP8 showed, by electron microscopy, the accumulation of tethered lysosome-associated membrane protein (LAMP)-carrier vesicles around multivesicular bodies, as well as the appearance of 'hourglass' profiles of late endocytic organelles attached by filamentous tethers, but did not prevent endocytic delivery to lysosomal hydrolases. Subsequent depletion of the SNARE YKT6 reduced this delivery, consistent with it compensating for the absence of VAMP7 and VAMP8. We also investigated filamentous tethering between multivesicular bodies and enlarged endolysosomes following depletion of charged multi-vesicular body protein 6 (CHMP6), and provide the first evidence that pore formation commences at the edge of tether arrays, with pore expansion required for full membrane fusion.


Subject(s)
Membrane Fusion , SNARE Proteins , Endosomes , HeLa Cells , Humans , Lysosomes , R-SNARE Proteins/genetics , SNARE Proteins/genetics
5.
Nat Commun ; 11(1): 5559, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33144569

ABSTRACT

Cholesterol import in mammalian cells is mediated by the LDL receptor pathway. Here, we perform a genome-wide CRISPR screen using an endogenous cholesterol reporter and identify >100 genes involved in LDL-cholesterol import. We characterise C18orf8 as a core subunit of the mammalian Mon1-Ccz1 guanidine exchange factor (GEF) for Rab7, required for complex stability and function. C18orf8-deficient cells lack Rab7 activation and show severe defects in late endosome morphology and endosomal LDL trafficking, resulting in cellular cholesterol deficiency. Unexpectedly, free cholesterol accumulates within swollen lysosomes, suggesting a critical defect in lysosomal cholesterol export. We find that active Rab7 interacts with the NPC1 cholesterol transporter and licenses lysosomal cholesterol export. This process is abolished in C18orf8-, Ccz1- and Mon1A/B-deficient cells and restored by a constitutively active Rab7. The trimeric Mon1-Ccz1-C18orf8 (MCC) GEF therefore plays a central role in cellular cholesterol homeostasis coordinating Rab7 activation, endosomal LDL trafficking and NPC1-dependent lysosomal cholesterol export.


Subject(s)
Cholesterol/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Protein Multimerization , rab GTP-Binding Proteins/metabolism , Biological Transport , CRISPR-Cas Systems/genetics , Cholesterol, LDL/metabolism , Endosomes/metabolism , Endosomes/ultrastructure , Fluorescent Dyes/metabolism , Genome, Human , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Hydroxymethylglutaryl-CoA Synthase/metabolism , Lysosomes/ultrastructure , Models, Biological , Multiprotein Complexes/metabolism , Niemann-Pick C1 Protein , Protein Binding , rab7 GTP-Binding Proteins
6.
Acta Neuropathol Commun ; 8(1): 165, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33059769

ABSTRACT

Autosomal dominant mutations in LITAF are responsible for the rare demyelinating peripheral neuropathy, Charcot-Marie-Tooth disease type 1C (CMT1C). The LITAF protein is expressed in many human cell types and we have investigated the consequences of two different LITAF mutations in primary fibroblasts from CMT1C patients using confocal and electron microscopy. We observed the appearance of vacuolation/enlargement of late endocytic compartments (late endosomes and lysosomes). This vacuolation was also observed after knocking out LITAF from either control human fibroblasts or from the CMT1C patient-derived cells, consistent with it being the result of loss-of-function mutations in the CMT1C fibroblasts. The vacuolation was similar to that previously observed in fibroblasts from CMT4J patients, which have autosomal recessive mutations in FIG4. The FIG4 protein is a component of a phosphoinositide kinase complex that synthesises phosphatidylinositol 3,5-bisphosphate on the limiting membrane of late endosomes. Phosphatidylinositol 3,5-bisphosphate activates the release of lysosomal Ca2+ through the cation channel TRPML1, which is required to maintain the homeostasis of endosomes and lysosomes in mammalian cells. We observed that a small molecule activator of TRPML1, ML-SA1, was able to rescue the vacuolation phenotype of LITAF knockout, FIG4 knockout and CMT1C patient fibroblasts. Our data describe the first cellular phenotype common to two different subtypes of demyelinating CMT and are consistent with LITAF and FIG4 functioning on a common endolysosomal pathway that is required to maintain the homeostasis of late endosomes and lysosomes. Although our experiments were on human fibroblasts, they have implications for our understanding of the molecular pathogenesis and approaches to therapy in two subtypes of demyelinating Charcot-Marie-Tooth disease.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , Lysosomes/metabolism , Transient Receptor Potential Channels/metabolism , Adult , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/physiopathology , Endosomes/pathology , Endosomes/ultrastructure , Female , Fibroblasts/pathology , Fibroblasts/ultrastructure , Flavoproteins/genetics , Gene Knockout Techniques , Humans , Loss of Function Mutation , Lysosomes/pathology , Lysosomes/ultrastructure , Male , Microscopy, Confocal , Microscopy, Electron , Middle Aged , Nuclear Proteins/genetics , Phosphoric Monoester Hydrolases/genetics , Transcription Factors/genetics , Vacuoles/pathology , Vacuoles/ultrastructure
7.
Nat Commun ; 11(1): 5031, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33024112

ABSTRACT

VARP and TBC1D5 are accessory/regulatory proteins of retromer-mediated retrograde trafficking from endosomes. Using an NMR/X-ray approach, we determined the structure of the complex between retromer subunit VPS29 and a 12 residue, four-cysteine/Zn++ microdomain, which we term a Zn-fingernail, two of which are present in VARP. Mutations that abolish VPS29:VARP binding inhibit trafficking from endosomes to the cell surface. We show that VARP and TBC1D5 bind the same site on VPS29 and can compete for binding VPS29 in vivo. The relative disposition of VPS29s in hetero-hexameric, membrane-attached, retromer arches indicates that VARP will prefer binding to assembled retromer coats through simultaneous binding of two VPS29s. The TBC1D5:VPS29 interaction is over one billion years old but the Zn-fingernail appears only in VARP homologues in the lineage directly giving rise to animals at which point the retromer/VARP/TBC1D5 regulatory network became fully established.


Subject(s)
Evolution, Molecular , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Zinc/metabolism , Cryoelectron Microscopy , Cysteine/chemistry , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Conformation , Vesicular Transport Proteins/genetics , Zinc Fingers
8.
Hum Mol Genet ; 28(15): 2514-2530, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31070736

ABSTRACT

A rare lysosomal disease resembling a mucopolysaccharidosis with unusual systemic features, including renal disease and platelet dysfunction, caused by the defect in a conserved region of the VPS33A gene on human chromosome 12q24.31, occurs in Yakuts-a nomadic Turkic ethnic group of Southern Siberia. VPS33A is a core component of the class C core vacuole/endosome tethering (CORVET) and the homotypic fusion and protein sorting (HOPS) complexes, which have essential functions in the endocytic pathway. Here we show that cultured fibroblasts from patients with this disorder have morphological changes: vacuolation with disordered endosomal/lysosomal compartments and-common to sphingolipid diseases-abnormal endocytic trafficking of lactosylceramide. Urine glycosaminoglycan studies revealed a pathological excess of sialylated conjugates as well as dermatan and heparan sulphate. Lipidomic screening showed elevated ß-D-galactosylsphingosine with unimpaired activity of cognate lysosomal hydrolases. The 3D crystal structure of human VPS33A predicts that replacement of arginine 498 by tryptophan will de-stabilize VPS33A folding. We observed that the missense mutation reduced the abundance of full-length VPS33A and other components of the HOPS and CORVET complexes. Treatment of HeLa cells stably expressing the mutant VPS33A with a proteasome inhibitor rescued the mutant protein from degradation. We propose that the disease is due to diminished intracellular abundance of intact VPS33A. Exposure of patient-derived fibroblasts to the clinically approved proteasome inhibitor, bortezomib, or inhibition of glucosylceramide synthesis with eliglustat, partially corrected the impaired lactosylceramide trafficking defect and immediately suggest therapeutic avenues to explore in this fatal orphan disease.


Subject(s)
Antigens, CD/metabolism , Carbohydrate Metabolism, Inborn Errors/genetics , Endocytosis , Lactosylceramides/metabolism , Lysosomes/metabolism , Mutation, Missense , Vesicular Transport Proteins/genetics , Bortezomib/therapeutic use , Carbohydrate Metabolism, Inborn Errors/metabolism , Carbohydrate Metabolism, Inborn Errors/physiopathology , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/pathology , HeLa Cells , Humans , Infant , Lysosomes/physiology , Male , Mucopolysaccharidoses , Phenotype , Proteasome Inhibitors/therapeutic use , Protein Conformation , Pyrrolidines/therapeutic use , Siberia , Vesicular Transport Proteins/metabolism , Exome Sequencing
9.
Nat Cell Biol ; 20(12): 1333-1335, 2018 12.
Article in English | MEDLINE | ID: mdl-30397316
10.
Prog Mol Subcell Biol ; 57: 151-180, 2018.
Article in English | MEDLINE | ID: mdl-30097775

ABSTRACT

In addition to being the terminal degradative compartment of the cell's endocytic and autophagic pathways, the lysosome is a multifunctional signalling hub integrating the cell's response to nutrient status and growth factor/hormone signalling. The cytosolic surface of the limiting membrane of the lysosome is the site of activation of the multiprotein complex mammalian target of rapamycin complex 1 (mTORC1), which phosphorylates numerous cell growth-related substrates, including transcription factor EB (TFEB). Under conditions in which mTORC1 is inhibited including starvation, TFEB becomes dephosphorylated and translocates to the nucleus where it functions as a master regulator of lysosome biogenesis. The signalling role of lysosomes is not limited to this pathway. They act as an intracellular Ca2+ store, which can release Ca2+ into the cytosol for both local effects on membrane fusion and pleiotropic effects within the cell. The relationship and crosstalk between the lysosomal and endoplasmic reticulum (ER) Ca2+ stores play a role in shaping intracellular Ca2+ signalling. Lysosomes also perform other signalling functions, which are discussed. Current views of the lysosomal compartment recognize its dynamic nature. It includes endolysosomes, autolysosome and storage lysosomes that are constantly engaged in fusion/fission events and lysosome regeneration. How signalling is affected by individual lysosomal organelles being at different stages of these processes and/or at different sites within the cell is poorly understood, but is discussed.


Subject(s)
Endocytosis/genetics , Endoplasmic Reticulum/genetics , Endosomes/genetics , Lysosomes/genetics , Animals , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Humans , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Signal Transduction/genetics
11.
J Cell Biol ; 214(3): 293-308, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27482051

ABSTRACT

Endomembrane organelle maturation requires cargo delivery via fusion with membrane transport intermediates and recycling of fusion factors to their sites of origin. Melanosomes and other lysosome-related organelles obtain cargoes from early endosomes, but the fusion machinery involved and its recycling pathway are unknown. Here, we show that the v-SNARE VAMP7 mediates fusion of melanosomes with tubular transport carriers that also carry the cargo protein TYRP1 and that require BLOC-1 for their formation. Using live-cell imaging, we identify a pathway for VAMP7 recycling from melanosomes that employs distinct tubular carriers. The recycling carriers also harbor the VAMP7-binding scaffold protein VARP and the tissue-restricted Rab GTPase RAB38. Recycling carrier formation is dependent on the RAB38 exchange factor BLOC-3. Our data suggest that VAMP7 mediates fusion of BLOC-1-dependent transport carriers with melanosomes, illuminate SNARE recycling from melanosomes as a critical BLOC-3-dependent step, and likely explain the distinct hypopigmentation phenotypes associated with BLOC-1 and BLOC-3 deficiency in Hermansky-Pudlak syndrome variants.


Subject(s)
Carrier Proteins/metabolism , Endocytosis , Lectins/metabolism , Melanosomes/metabolism , Nerve Tissue Proteins/metabolism , R-SNARE Proteins/metabolism , Transport Vesicles/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Melanocytes/metabolism , Melanocytes/ultrastructure , Melanosomes/ultrastructure , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mitochondrial Proteins , Oxidoreductases/metabolism , Pigmentation , Protein Transport , Qa-SNARE Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transport Vesicles/ultrastructure , rab GTP-Binding Proteins/metabolism
12.
Curr Biol ; 26(17): 2233-45, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27498570

ABSTRACT

The endocytic delivery of macromolecules from the mammalian cell surface for degradation by lysosomal acid hydrolases requires traffic through early endosomes to late endosomes followed by transient (kissing) or complete fusions between late endosomes and lysosomes. Transient or complete fusion results in the formation of endolysosomes, which are hybrid organelles from which lysosomes are re-formed. We have used synthetic membrane-permeable cathepsin substrates, which liberate fluorescent reporters upon proteolytic cleavage, as well as acid phosphatase cytochemistry to identify which endocytic compartments are acid hydrolase active. We found that endolysosomes are the principal organelles in which acid hydrolase substrates are cleaved. Endolysosomes also accumulated acidotropic probes and could be distinguished from terminal storage lysosomes, which were acid hydrolase inactive and did not accumulate acidotropic probes. Using live-cell microscopy, we have demonstrated that fusion events, which form endolysosomes, precede the onset of acid hydrolase activity. By means of sucrose and invertase uptake experiments, we have also shown that acid-hydrolase-active endolysosomes and acid-hydrolase-inactive, terminal storage lysosomes exist in dynamic equilibrium. We conclude that the terminal endocytic compartment is composed of acid-hydrolase-active, acidic endolysosomes and acid hydrolase-inactive, non-acidic, terminal storage lysosomes, which are linked and function in a lysosome regeneration cycle.


Subject(s)
Endosomes/metabolism , Hydrolases/metabolism , Lysosomes/metabolism , Fibroblasts , HeLa Cells , Humans , MCF-7 Cells
13.
Traffic ; 17(8): 940-58, 2016 08.
Article in English | MEDLINE | ID: mdl-27126989

ABSTRACT

Tetherin (BST2/CD317) is a viral restriction factor that anchors enveloped viruses to host cells and limits viral spread. The HIV-1 Vpu accessory protein counteracts tetherin by decreasing its cell surface expression and targeting it for ubiquitin-dependent endolysosomal degradation. Although the Vpu-mediated downregulation of tetherin has been extensively studied, the molecular details are not completely elucidated. We therefore used a forward genetic screen in human haploid KBM7 cells to identify novel genes required for tetherin trafficking. Our screen identified WDR81 as a novel gene required for tetherin trafficking and degradation in both the presence and absence of Vpu. WDR81 is a BEACH-domain containing protein that is also required for the degradation of EGF-stimulated epidermal growth factor receptor (EGFR) and functions in a complex with the WDR91 protein. In the absence of WDR81 the endolysosomal compartment appears swollen, with enlarged early and late endosomes and reduced delivery of endocytosed dextran to cathepsin-active lysosomes. Our data suggest a role for the WDR81-WDR91 complex in the fusion of endolysosomal compartments and the absence of WDR81 leads to impaired receptor trafficking and degradation.


Subject(s)
Antigens, CD/metabolism , Carrier Proteins/metabolism , Lysosomes/metabolism , Nerve Tissue Proteins/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , GPI-Linked Proteins/metabolism , HIV-1/metabolism , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Humans , Protein Transport , Viral Regulatory and Accessory Proteins/genetics
14.
Biochem J ; 471(1): 79-88, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26221024

ABSTRACT

The Kaposi's sarcoma-associated herpes virus (KSHV) K3 viral gene product effectively down-regulates cell surface MHC class I. K3 is an E3 ubiquitin ligase that promotes Lys(63)-linked polyubiquitination of MHC class I, providing the signal for clathrin-mediated endocytosis. Endocytosis is followed by sorting into the intralumenal vesicles (ILVs) of multivesicular bodies (MVBs) and eventual delivery to lysosomes. The sorting of MHC class I into MVBs requires many individual proteins of the four endosomal sorting complexes required for transport (ESCRTs). In HeLa cells expressing the KSHV K3 ubiquitin ligase, the effect of RNAi-mediated depletion of individual proteins of the ESCRT-0 and ESCRT-I complexes and three ESCRT-III proteins showed that these are required to down-regulate MHC class I. However, depletion of proteins of the ESCRT-II complex or of the ESCRT-III protein, VPS20 (vacuolar protein sorting 20)/CHMP6 (charged MVB protein 6), failed to prevent the loss of MHC class I from the cell surface. Depletion of histidine domain phosphotyrosine phosphatase (HD-PTP) resulted in an increase in the cell surface concentration of MHC class I in HeLa cells expressing the KSHV K3 ubiquitin ligase. Rescue experiments with wild-type (WT) and mutant HD-PTP supported the conclusion that HD-PTP acts as an alternative to ESCRT-II and VPS20/CHMP6 as a link between the ESCRT-I and those ESCRT-III protein(s) necessary for ILV formation. Thus, the down-regulation of cell surface MHC class I, polyubiquitinated by the KSHV K3 ubiquitin ligase, does not employ the canonical ESCRT pathway, but instead utilizes an alternative pathway in which HD-PTP replaces ESCRT-II and VPS20/CHMP6.


Subject(s)
Down-Regulation , Endosomal Sorting Complexes Required for Transport/metabolism , Herpesvirus 8, Human/metabolism , Histocompatibility Antigens Class I/biosynthesis , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Ubiquitination , Viral Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HeLa Cells , Herpesvirus 8, Human/genetics , Histocompatibility Antigens Class I/genetics , Humans , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Viral Proteins/genetics
15.
Curr Biol ; 25(8): R315-6, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25898096
16.
Traffic ; 16(7): 727-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25783203

ABSTRACT

The mammalian homotypic fusion and vacuole protein sorting (HOPS) complex is comprised of six subunits: VPS11, VPS16, VPS18, VPS39, VPS41 and the Sec1/Munc18 (SM) family member VPS33A. Human HOPS has been predicted to be a tethering complex required for fusion of intracellular compartments with lysosomes, but it remains unclear whether all HOPS subunits are required. We showed that the whole HOPS complex is required for fusion of endosomes with lysosomes by monitoring the delivery of endocytosed fluorescent dextran to lysosomes in cells depleted of individual HOPS proteins. We used the crystal structure of the VPS16/VPS33A complex to design VPS16 and VPS33A mutants that no longer bind each other and showed that, unlike the wild-type proteins, these mutants no longer rescue lysosome fusion with endosomes or autophagosomes in cells depleted of the endogenous proteins. There was no effect of depleting either VIPAR or VPS33B, paralogs of VPS16 and VPS33A, on fusion of lysosomes with either endosomes or autophagosomes and immunoprecipitation showed that they form a complex distinct from HOPS. Our data demonstrate the necessity of recruiting the SM protein VPS33A to HOPS via its interaction with VPS16 and that HOPS proteins, but not VIPAR or VPS33B, are essential for fusion of endosomes or autophagosomes with lysosomes.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Phagosomes/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Autophagy , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Transport , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
17.
Methods Cell Biol ; 126: 101-18, 2015.
Article in English | MEDLINE | ID: mdl-25665443

ABSTRACT

In mammalian cells, lysosomes fuse with late endosomes to form endolysosomes from which lysosomes are reformed. Lysosomal fusion events were initially inferred from light and electron microscopy studies, demonstrated in cell-free content mixing assays and, more recently, shown directly with live cell microscopy. Currently, there is a focus on studying lysosome fusion in cultured cells using various forms of microscopy, especially under conditions in which the use of overexpression of dominant-negative protein constructs or the use of RNA interference to deplete individual proteins allows the investigation of the molecular machinery of fusion. Here, we review a variety of fluorescence, live cell, and electron microscopy techniques with which to study lysosome fusion in cultured mammalian cells. We address the merits and limitations of different techniques when choosing an assay system and provide a series of protocols with which to study endocytic delivery to lysosomes and fusion events between lysosomes and endosomes.


Subject(s)
Lysosomes/metabolism , Caco-2 Cells , Endocytosis , Endosomes/metabolism , Fluorescent Dyes/chemistry , HeLa Cells , Humans , MCF-7 Cells , Membrane Fusion , Microscopy, Electron, Transmission , Microscopy, Fluorescence
18.
Cold Spring Harb Perspect Biol ; 6(9): a016840, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25183830

ABSTRACT

Lysosomes were once considered the end point of endocytosis, simply used for macromolecule degradation. They are now recognized to be dynamic organelles, able to fuse with a variety of targets and to be re-formed after fusion events. They are also now known to be the site of nutrient sensing and signaling to the cell nucleus. In addition, lysosomes are secretory organelles, with specialized machinery for regulated secretion of proteins in some cell types. The biogenesis of lysosomes and lysosome-related organelles is discussed, taking into account their dynamic nature and multiple roles.


Subject(s)
Lysosomes/metabolism , Organelles/metabolism , Amino Acid Motifs , Animals , Apoptosis , Autophagy , Biological Transport , Endocytosis , Endosomes/metabolism , Humans , Immune System , Killer Cells, Natural/cytology , SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/cytology , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins
19.
Dev Cell ; 29(5): 591-606, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24856514

ABSTRACT

VARP is a Rab32/38 effector that also binds to the endosomal/lysosomal R-SNARE VAMP7. VARP binding regulates VAMP7 participation in SNARE complex formation and can therefore influence VAMP7-mediated membrane fusion events. Mutant versions of VARP that cannot bind Rab32:GTP, designed on the basis of the VARP ankyrin repeat/Rab32:GTP complex structure described here, unexpectedly retain endosomal localization, showing that VARP recruitment is not dependent on Rab32 binding. We show that recruitment of VARP to the endosomal membrane is mediated by its direct interaction with VPS29, a subunit of the retromer complex, which is involved in trafficking from endosomes to the TGN and the cell surface. Transport of GLUT1 from endosomes to the cell surface requires VARP, VPS29, and VAMP7 and depends on the direct interaction between VPS29 and VARP. Finally, we propose that endocytic cycling of VAMP7 depends on its interaction with VARP and, consequently, also on retromer.


Subject(s)
Cell Membrane/metabolism , Endosomes/physiology , Glucose Transporter Type 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism , R-SNARE Proteins/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Amino Acid Sequence , Blotting, Western , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Immunoprecipitation , Molecular Sequence Data , Muscle Proteins/metabolism , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Protein Transport , R-SNARE Proteins/chemistry , R-SNARE Proteins/genetics , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
20.
Proc Natl Acad Sci U S A ; 110(33): 13345-50, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901104

ABSTRACT

The multisubunit homotypic fusion and vacuole protein sorting (HOPS) membrane-tethering complex is required for late endosome-lysosome and autophagosome-lysosome fusion in mammals. We have determined the crystal structure of the human HOPS subunit Vps33A, confirming its identity as a Sec1/Munc18 family member. We show that HOPS subunit Vps16 recruits Vps33A to the human HOPS complex and that residues 642-736 are necessary and sufficient for this interaction, and we present the crystal structure of Vps33A in complex with Vps16(642-736). Mutations at the binding interface disrupt the Vps33A-Vps16 interaction both in vitro and in cells, preventing recruitment of Vps33A to the HOPS complex. The Vps33A-Vps16 complex provides a structural framework for studying the association between Sec1/Munc18 proteins and tethering complexes.


Subject(s)
Models, Molecular , Multiprotein Complexes/chemistry , Protein Conformation , Vesicular Transport Proteins/chemistry , Binding Sites/genetics , Escherichia coli , Humans , Multiprotein Complexes/metabolism , Mutation/genetics , Species Specificity , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...