Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 701
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 182(1): 98-111.e18, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32544384

ABSTRACT

Lysosomal cholesterol egress requires two proteins, NPC1 and NPC2, whose defects are responsible for Niemann-Pick disease type C (NPC). Here, we present systematic structural characterizations that reveal the molecular basis for low-pH-dependent cholesterol delivery from NPC2 to the transmembrane (TM) domain of NPC1. At pH 8.0, similar structures of NPC1 were obtained in nanodiscs and in detergent at resolutions of 3.6 Å and 3.0 Å, respectively. A tunnel connecting the N-terminal domain (NTD) and the transmembrane sterol-sensing domain (SSD) was unveiled. At pH 5.5, the NTD exhibits two conformations, suggesting the motion for cholesterol delivery to the tunnel. A putative cholesterol molecule is found at the membrane boundary of the tunnel, and TM2 moves toward formation of a surface pocket on the SSD. Finally, the structure of the NPC1-NPC2 complex at 4.0 Å resolution was obtained at pH 5.5, elucidating the molecular basis for cholesterol handoff from NPC2 to NPC1(NTD).


Subject(s)
Cholesterol/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Green Fluorescent Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Models, Molecular , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Niemann-Pick C1 Protein , Protein Domains , Structural Homology, Protein , Structure-Activity Relationship
2.
Cell ; 179(2): 485-497.e18, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31543266

ABSTRACT

Niemann-Pick type C (NPC) proteins are essential for sterol homeostasis, believed to drive sterol integration into the lysosomal membrane before redistribution to other cellular membranes. Here, using a combination of crystallography, cryo-electron microscopy, and biochemical and in vivo studies on the Saccharomyces cerevisiae NPC system (NCR1 and NPC2), we present a framework for sterol membrane integration. Sterols are transferred between hydrophobic pockets of vacuolar NPC2 and membrane-protein NCR1. NCR1 has its N-terminal domain (NTD) positioned to deliver a sterol to a tunnel connecting NTD to the luminal membrane leaflet 50 Å away. A sterol is caught inside this tunnel during transport, and a proton-relay network of charged residues in the transmembrane region is linked to this tunnel supporting a proton-driven transport mechanism. We propose a model for sterol integration that clarifies the role of NPC proteins in this essential eukaryotic pathway and that rationalizes mutations in patients with Niemann-Pick disease type C.


Subject(s)
Carrier Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Sterols/metabolism , Vesicular Transport Proteins/chemistry , Biological Transport , Cryoelectron Microscopy , Crystallography , Intracellular Membranes/metabolism , Lysosomes/metabolism , Protein Domains , Vacuoles/metabolism
3.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348885

ABSTRACT

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Subject(s)
Benzamides/metabolism , Bridged Bicyclo Compounds/pharmacology , Heptanes/pharmacology , Lysosomes/drug effects , Vesicular Transport Proteins/metabolism , Activating Transcription Factor 6/metabolism , Animals , Benzamides/chemistry , Benzamides/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Frameshift Mutation , Heptanes/therapeutic use , Humans , Imidazoline Receptors/antagonists & inhibitors , Imidazoline Receptors/genetics , Imidazoline Receptors/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lysosomes/metabolism , Male , Mice , Mice, Transgenic , Mucin-1/chemistry , Mucin-1/genetics , Mucin-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Unfolded Protein Response/drug effects , Vesicular Transport Proteins/chemistry
4.
Cell ; 171(2): 346-357.e12, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28919078

ABSTRACT

Newly synthesized proteins engage molecular chaperones that assist folding. Their progress is monitored by quality control systems that target folding errors for degradation. Paradoxically, chaperones that promote folding also direct unfolded polypeptides for degradation. Hence, a mechanism was previously hypothesized that prevents the degradation of actively folding polypeptides. In this study, we show that a conserved endoplasmic reticulum (ER) membrane protein complex, consisting of Slp1 and Emp65 proteins, performs this function in the ER lumen. The complex binds unfolded proteins and protects them from degradation during folding. In its absence, approximately 20%-30% of newly synthesized proteins that could otherwise fold are degraded. Although the Slp1-Emp65 complex hosts a broad range of clients, it is specific for soluble proteins. Taken together, these studies demonstrate the vulnerability of newly translated, actively folding polypeptides and the discovery of a new proteostasis functional class we term "guardian" that protects them from degradation.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins/metabolism , Animals , Endoplasmic Reticulum-Associated Degradation , Glycosylation , Mice , Molecular Chaperones/metabolism , Proteolysis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/chemistry , Vesicular Transport Proteins/chemistry
5.
Cell ; 167(6): 1452-1454, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912055

ABSTRACT

Membrane trafficking from endosomes to the trans-Golgi network or the plasma membrane is driven by the retromer complex. Through structural analysis of the cargo-bound complex, Lucas et al. describe a mechanism by which endosomal membrane recruitment and cargo recognition are integrated through cooperative interactions between retromer subunits.


Subject(s)
Endosomes/metabolism , trans-Golgi Network/metabolism , Cell Membrane/metabolism , Humans , Intracellular Membranes/metabolism , Protein Transport , Vesicular Transport Proteins/chemistry
6.
Cell ; 167(6): 1623-1635.e14, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27889239

ABSTRACT

Retromer is a multi-protein complex that recycles transmembrane cargo from endosomes to the trans-Golgi network and the plasma membrane. Defects in retromer impair various cellular processes and underlie some forms of Alzheimer's disease and Parkinson's disease. Although retromer was discovered over 15 years ago, the mechanisms for cargo recognition and recruitment to endosomes have remained elusive. Here, we present an X-ray crystallographic analysis of a four-component complex comprising the VPS26 and VPS35 subunits of retromer, the sorting nexin SNX3, and a recycling signal from the divalent cation transporter DMT1-II. This analysis identifies a binding site for canonical recycling signals at the interface between VPS26 and SNX3. In addition, the structure highlights a network of cooperative interactions among the VPS subunits, SNX3, and cargo that couple signal-recognition to membrane recruitment.


Subject(s)
Cation Transport Proteins/chemistry , Multiprotein Complexes/chemistry , Sorting Nexins/chemistry , Vesicular Transport Proteins/chemistry , Amino Acid Sequence , Cation Transport Proteins/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Scattering, Small Angle , Sorting Nexins/metabolism , Vesicular Transport Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 121(33): e2405041121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39116126

ABSTRACT

Endosomal membrane trafficking is mediated by specific protein coats and formation of actin-rich membrane domains. The Retromer complex coordinates with sorting nexin (SNX) cargo adaptors including SNX27, and the SNX27-Retromer assembly interacts with the Wiskott-Aldrich syndrome protein and SCAR homolog (WASH) complex which nucleates actin filaments establishing the endosomal recycling domain. Crystal structures, modeling, biochemical, and cellular validation reveal how the FAM21 subunit of WASH interacts with both Retromer and SNX27. FAM21 binds the FERM domain of SNX27 using acidic-Asp-Leu-Phe (aDLF) motifs similar to those found in the SNX1 and SNX2 subunits of the ESCPE-1 complex. Overlapping FAM21 repeats and a specific Pro-Leu containing motif bind three distinct sites on Retromer involving both the VPS35 and VPS29 subunits. Mutation of the major VPS35-binding site does not prevent cargo recycling; however, it partially reduces endosomal WASH association indicating that a network of redundant interactions promote endosomal activity of the WASH complex. These studies establish the molecular basis for how SNX27-Retromer is coupled to the WASH complex via overlapping and multiplexed motif-based interactions required for the dynamic assembly of endosomal membrane recycling domains.


Subject(s)
Endosomes , Sorting Nexins , Vesicular Transport Proteins , Humans , Endosomes/metabolism , Sorting Nexins/metabolism , Sorting Nexins/genetics , Sorting Nexins/chemistry , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/chemistry , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/chemistry , Protein Binding , Crystallography, X-Ray , Binding Sites , Models, Molecular
8.
EMBO J ; 40(12): e107607, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34018207

ABSTRACT

The GTPase Rab1 is a master regulator of the early secretory pathway and is critical for autophagy. Rab1 activation is controlled by its guanine nucleotide exchange factor, the multisubunit TRAPPIII complex. Here, we report the 3.7 Å cryo-EM structure of the Saccharomyces cerevisiae TRAPPIII complex bound to its substrate Rab1/Ypt1. The structure reveals the binding site for the Rab1/Ypt1 hypervariable domain, leading to a model for how the complex interacts with membranes during the activation reaction. We determined that stable membrane binding by the TRAPPIII complex is required for robust activation of Rab1/Ypt1 in vitro and in vivo, and is mediated by a conserved amphipathic α-helix within the regulatory Trs85 subunit. Our results show that the Trs85 subunit serves as a membrane anchor, via its amphipathic helix, for the entire TRAPPIII complex. These findings provide a structural understanding of Rab activation on organelle and vesicle membranes.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Vesicular Transport Proteins/chemistry , rab GTP-Binding Proteins/chemistry , Cryoelectron Microscopy , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Protein Conformation , Saccharomyces cerevisiae Proteins/ultrastructure , Vesicular Transport Proteins/ultrastructure , rab GTP-Binding Proteins/ultrastructure
9.
EMBO J ; 40(12): e107608, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34018214

ABSTRACT

The TRAPP complexes are nucleotide exchange factors that play essential roles in membrane traffic and autophagy. TRAPPII activates Rab11, and TRAPPIII activates Rab1, with the two complexes sharing a core of small subunits that affect nucleotide exchange but being distinguished by specific large subunits that are essential for activity in vivo. Crystal structures of core subunits have revealed the mechanism of Rab activation, but how the core and the large subunits assemble to form the complexes is unknown. We report a cryo-EM structure of the entire Drosophila TRAPPIII complex. The TRAPPIII-specific subunits TRAPPC8 and TRAPPC11 hold the catalytic core like a pair of tongs, with TRAPPC12 and TRAPPC13 positioned at the joint between them. TRAPPC2 and TRAPPC2L link the core to the two large arms, with the interfaces containing residues affected by disease-causing mutations. The TRAPPC8 arm is positioned such that it would contact Rab1 that is bound to the core, indicating how the arm could determine the specificity of the complex. A lower resolution structure of TRAPPII shows a similar architecture and suggests that the TRAPP complexes evolved from a single ur-TRAPP.


Subject(s)
Drosophila Proteins/chemistry , Vesicular Transport Proteins/chemistry , rab1 GTP-Binding Proteins/chemistry , Cryoelectron Microscopy , Drosophila Proteins/ultrastructure , Guanine Nucleotide Exchange Factors/chemistry , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Protein Conformation , Vesicular Transport Proteins/ultrastructure , rab1 GTP-Binding Proteins/ultrastructure
10.
EMBO J ; 39(23): e104369, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33124732

ABSTRACT

Organelles are physically connected in membrane contact sites. The endoplasmic reticulum possesses three major receptors, VAP-A, VAP-B, and MOSPD2, which interact with proteins at the surface of other organelles to build contacts. VAP-A, VAP-B, and MOSPD2 contain an MSP domain, which binds a motif named FFAT (two phenylalanines in an acidic tract). In this study, we identified a non-conventional FFAT motif where a conserved acidic residue is replaced by a serine/threonine. We show that phosphorylation of this serine/threonine is critical for non-conventional FFAT motifs (named Phospho-FFAT) to be recognized by the MSP domain. Moreover, structural analyses of the MSP domain alone or in complex with conventional and Phospho-FFAT peptides revealed new mechanisms of interaction. Based on these new insights, we produced a novel prediction algorithm, which expands the repertoire of candidate proteins with a Phospho-FFAT that are able to create membrane contact sites. Using a prototypical tethering complex made by STARD3 and VAP, we showed that phosphorylation is instrumental for the formation of ER-endosome contacts, and their sterol transfer function. This study reveals that phosphorylation acts as a general switch for inter-organelle contacts.


Subject(s)
Lipid Metabolism , Membrane Proteins/metabolism , Receptors, Chemokine/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Binding Sites , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Humans , Lipids , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Phosphorylation , Protein Binding , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
11.
Plant Cell ; 33(2): 404-419, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33630076

ABSTRACT

During the immune response, activation of the secretory pathway is key to mounting an effective response, while gauging its output is important to maintain cellular homeostasis. The Exo70 subunit of the exocyst functions as a spatiotemporal regulator by mediating numerous interactions with proteins and lipids. However, a molecular understanding of the exocyst regulation remains challenging. We show that, in Arabidopsis thaliana, Exo70B2 behaves as a bona fide exocyst subunit. Conversely, treatment with the salicylic acid (SA) defence hormone analog benzothiadiazole (BTH), or the immunogenic peptide flg22, induced Exo70B2 transport into the vacuole. We reveal that Exo70B2 interacts with AUTOPHAGY-RELATED PROTEIN 8 (ATG8) via two ATG8-interacting motives (AIMs) and its transport into the vacuole is dependent on autophagy. In line with its role in immunity, we discovered that Exo70B2 interacted with and was phosphorylated by the kinase MPK3. Mimicking phosphorylation had a dual impact on Exo70B2: first, by inhibiting localization at sites of active secretion, and second, it increased the interaction with ATG8. Phosphonull variants displayed higher effector-triggered immunity (ETI) and were hypersensitive to BTH, which induce secretion and autophagy. Our results suggest a molecular mechanism by which phosphorylation diverts Exo70B2 from the secretory into the autophagy pathway for its degradation, to dampen secretory activity.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Autophagy/immunology , Protein Subunits/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Arabidopsis/drug effects , Arabidopsis/microbiology , Arabidopsis Proteins/chemistry , Autophagy/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Pseudomonas syringae/drug effects , Pseudomonas syringae/physiology , Signal Transduction/drug effects , Thiadiazoles/pharmacology , Vacuoles/drug effects , Vacuoles/metabolism , Vesicular Transport Proteins/chemistry , Virulence/drug effects , trans-Golgi Network/drug effects , trans-Golgi Network/metabolism
12.
Cell ; 136(1): 15-7, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19135881

ABSTRACT

The ESCRT-III complex mediates membrane budding away from the cytosol in endosome biogenesis, cytokinesis, and viral budding. In this issue, Saksena et al. (2009) use an elegant fluorescence-based approach to define the sequential activation, recruitment, and disassembly of ESCRT-III subunits during membrane involution in vitro.


Subject(s)
Endosomes/chemistry , Endosomes/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Cytosol/chemistry , Cytosol/metabolism , Humans , Spectrometry, Fluorescence , Yeasts
13.
Cell ; 136(1): 97-109, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19135892

ABSTRACT

Receptor downregulation in the MVB pathway is mediated by the ESCRT complexes. ESCRT-III is composed of four protein subunits that are monomeric in the cytosol and oligomerize into a protein lattice only upon membrane binding. Recent studies have shown that the ESCRT-III protein Snf7 can form a filament by undergoing homo-oligomerization. To examine the role of membrane binding and of interactions with other ESCRT components in initiating Snf7 oligomerization, we used fluorescence spectroscopy to directly detect and characterize the assembly of the Snf7 oligomer on liposomes using purified ESCRT components. The observed fluorescence changes reveal an obligatory sequence of membrane-protein and protein-protein interactions that generate the active conformation of Snf7. Also, we demonstrate that ESCRT-III assembly drives membrane deformation. Furthermore, using an in vitro disassembly assay, we directly demonstrate that Vps24 and Vps2 function as adaptors in the ATP-dependent membrane disassembly of the ESCRT-III complex by recruiting the AAA ATPase Vps4.


Subject(s)
Endosomes/chemistry , Endosomes/metabolism , Spectrometry, Fluorescence , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Endosomal Sorting Complexes Required for Transport , Humans , Liposomes/chemistry , Liposomes/metabolism , Multiprotein Complexes/metabolism , Yeasts
14.
Nature ; 561(7724): 561-564, 2018 09.
Article in English | MEDLINE | ID: mdl-30224749

ABSTRACT

Eukaryotic cells traffic proteins and lipids between different compartments using protein-coated vesicles and tubules. The retromer complex is required to generate cargo-selective tubulovesicular carriers from endosomal membranes1-3. Conserved in eukaryotes, retromer controls the cellular localization and homeostasis of hundreds of transmembrane proteins, and its disruption is associated with major neurodegenerative disorders4-7. How retromer is assembled and how it is recruited to form coated tubules is not known. Here we describe the structure of the retromer complex (Vps26-Vps29-Vps35) assembled on membrane tubules with the bin/amphiphysin/rvs-domain-containing sorting nexin protein Vps5, using cryo-electron tomography and subtomogram averaging. This reveals a membrane-associated Vps5 array, from which arches of retromer extend away from the membrane surface. Vps35 forms the 'legs' of these arches, and Vps29 resides at the apex where it is free to interact with regulatory factors. The bases of the arches connect to each other and to Vps5 through Vps26, and the presence of the same arches on coated tubules within cells confirms their functional importance. Vps5 binds to Vps26 at a position analogous to the previously described cargo- and Snx3-binding site, which suggests the existence of distinct retromer-sorting nexin assemblies. The structure provides insight into the architecture of the coat and its mechanism of assembly, and suggests that retromer promotes tubule formation by directing the distribution of sorting nexin proteins on the membrane surface while providing a scaffold for regulatory-protein interactions.


Subject(s)
Chaetomium/chemistry , Chaetomium/ultrastructure , Cryoelectron Microscopy , Electron Microscope Tomography , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/ultrastructure , Chaetomium/metabolism , Chlamydomonas reinhardtii/cytology , Chlamydomonas reinhardtii/ultrastructure , Humans , Models, Molecular , Protein Binding , Protein Transport , Sorting Nexins/chemistry , Sorting Nexins/metabolism , Sorting Nexins/ultrastructure , Vesicular Transport Proteins/metabolism
15.
J Biol Chem ; 298(5): 101780, 2022 05.
Article in English | MEDLINE | ID: mdl-35231443

ABSTRACT

Membrane contact sites are specialized areas where the membranes of two distinct organelles are physically connected and allow for the exchange of molecules and for signaling processes. Understanding the mechanisms whereby proteins localize to and function in these structures is of special interest; however, methods allowing for reconstitution of these contact sites are few and only based on synthetic membranes and recombinant proteins. Here, we devised a strategy to create in situ artificial contact sites between synthetic and endogenous organelle membranes. Liposomes functionalized with a peptide containing a two phenylalanines in an acidic tract (FFAT) motif were added to adherent cells whose plasma membrane was perforated. Confocal and super-resolution microscopy revealed that these liposomes associated with the endoplasmic reticulum via the specific interaction of the FFAT motif with endoplasmic reticulum-resident vesicle-associated membrane protein-associated proteins. This approach allowed for quantification of the attachment properties of peptides corresponding to FFAT motifs derived from distinct proteins and of a protein construct derived from steroidogenic acute regulatory protein-related lipid transfer domain-3. Collectively, these data indicate that the creation of in situ artificial contact sites represents an efficient approach for studying the membrane-tethering activity of proteins and for designing membrane contact site reconstitution assays in cellular contexts.


Subject(s)
Endoplasmic Reticulum , Liposomes , Membranes, Artificial , Amino Acid Motifs , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Liposomes/chemistry , Liposomes/metabolism , Liposomes/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombinant Proteins , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
16.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33053334

ABSTRACT

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/metabolism , HSP47 Heat-Shock Proteins/metabolism , Osteogenesis Imperfecta/genetics , Vesicular Transport Proteins/metabolism , Adult , Alleles , Amino Acid Sequence , Animals , Binding Sites , Bone and Bones/pathology , Chickens , Child, Preschool , Collagen Type I/chemistry , Collagen Type I/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , Humans , Infant , Male , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Pedigree , Primary Cell Culture , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
17.
PLoS Pathog ; 17(11): e1009409, 2021 11.
Article in English | MEDLINE | ID: mdl-34843601

ABSTRACT

The HIV-1 accessory protein Vpu modulates membrane protein trafficking and degradation to provide evasion of immune surveillance. Targets of Vpu include CD4, HLAs, and BST-2. Several cellular pathways co-opted by Vpu have been identified, but the picture of Vpu's itinerary and activities within membrane systems remains incomplete. Here, we used fusion proteins of Vpu and the enzyme ascorbate peroxidase (APEX2) to compare the ultrastructural locations and the proximal proteomes of wild type Vpu and Vpu-mutants. The proximity-omes of the proteins correlated with their ultrastructural locations and placed wild type Vpu near both retromer and ESCRT-0 complexes. Hierarchical clustering of protein abundances across the mutants was essential to interpreting the data and identified Vpu degradation-targets including CD4, HLA-C, and SEC12 as well as Vpu-cofactors including HGS, STAM, clathrin, and PTPN23, an ALIX-like protein. The Vpu-directed degradation of BST-2 was supported by STAM and PTPN23 and to a much lesser extent by the retromer subunits Vps35 and SNX3. PTPN23 also supported the Vpu-directed decrease in CD4 at the cell surface. These data suggest that Vpu directs targets from sorting endosomes to degradation at multi-vesicular bodies via ESCRT-0 and PTPN23.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , HIV Infections/virology , Human Immunodeficiency Virus Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proteome/metabolism , Sorting Nexins/metabolism , Vesicular Transport Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Viroporin Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/physiology , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Humans , Microscopy, Electron , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Proteome/analysis , Sorting Nexins/chemistry , Sorting Nexins/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Viroporin Proteins/genetics
18.
Cell ; 134(3): 384-5, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18692460

ABSTRACT

Anterograde transport in the early secretory pathway is mediated by COPII-coated vesicles. Stagg et al. (2008) have now visualized the double-layered COPII coat using electron cryomicroscopy, providing insight into how coats are assembled to accommodate cargo of different sizes.


Subject(s)
Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Transport
19.
Cell ; 134(3): 474-84, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18692470

ABSTRACT

Using cryo-electron microscopy, we have solved the structure of an icosidodecahedral COPII coat involved in cargo export from the endoplasmic reticulum (ER) coassembled from purified cargo adaptor Sec23-24 and Sec13-31 lattice-forming complexes. The coat structure shows a tetrameric assembly of the Sec23-24 adaptor layer that is well positioned beneath the vertices and edges of the Sec13-31 lattice. Fitting the known crystal structures of the COPII proteins into the density map reveals a flexible hinge region stemming from interactions between WD40 beta-propeller domains present in Sec13 and Sec31 at the vertices. The structure shows that the hinge region can direct geometric cage expansion to accommodate a wide range of bulky cargo, including procollagen and chylomicrons, that is sensitive to adaptor function in inherited disease. The COPII coat structure leads us to propose a mechanism by which cargo drives cage assembly and membrane curvature for budding from the ER.


Subject(s)
Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Transport
20.
Cell ; 133(7): 1202-13, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18585354

ABSTRACT

The multimeric membrane-tethering complexes TRAPPI and TRAPPII share seven subunits, of which four (Bet3p, Bet5p, Trs23p, and Trs31p) are minimally needed to activate the Rab GTPase Ypt1p in an event preceding membrane fusion. Here, we present the structure of a heteropentameric TRAPPI assembly complexed with Ypt1p. We propose that TRAPPI facilitates nucleotide exchange primarily by stabilizing the nucleotide-binding pocket of Ypt1p in an open, solvent-accessible form. Bet3p, Bet5p, and Trs23p interact directly with Ypt1p to stabilize this form, while the C terminus of Bet3p invades the pocket to participate in its remodeling. The Trs31p subunit does not interact directly with the GTPase but allosterically regulates the TRAPPI interface with Ypt1p. Our findings imply that TRAPPII activates Ypt1p by an identical mechanism. This view of a multimeric membrane-tethering assembly complexed with a Rab provides a framework for understanding events preceding membrane fusion at the molecular level.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Enzyme Activation , Golgi Apparatus/metabolism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Models, Molecular , Protein Interaction Mapping , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL