Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 698
Filter
1.
Stud Health Technol Inform ; 308: 351-358, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38007759

ABSTRACT

In eukaryotic cells, vesicular transport plays a crucial role in the docking and fusion of secretory vesicles with their respective target membranes. This intricate process is dependent on a complex network of multiple molecules. One of the important processes is tethering. The exocyst complex facilitates the tethering of secretory vesicles to the plasma membrane during exocytosis. The Sec6 subunit in yeast interacts with other exocyst subunits and may regulate SNARE assembly, which is crucial for understanding the assembly mechanism of exocyst and its interaction with SNARE. In this study, we designed two truncated forms of HuSec6, HuSec6 121-734 and HuSec6 121-745, based on results of bioinformatics analysis. We expressed and purified the proteins in E. coli, obtaining a protein purity of over 95% and protein crystals. X-ray diffraction results showed a resolution of approximately 9 Å for the crystals, providing a solid foundation for the crystal structure analysis of HuSec6.


Subject(s)
Escherichia coli , Vesicular Transport Proteins , Humans , Escherichia coli/metabolism , Exocytosis/physiology , Saccharomyces cerevisiae/metabolism , SNARE Proteins/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
2.
Nat Commun ; 14(1): 1768, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997511

ABSTRACT

The energy sensor AMP-activated protein kinase (AMPK) can activate autophagy when cellular energy production becomes compromised. However, the degree to which nutrient sensing impinges on the autophagosome closure remains unknown. Here, we provide the mechanism underlying a plant unique protein FREE1, upon autophagy-induced SnRK1α1-mediated phosphorylation, functions as a linkage between ATG conjugation system and ESCRT machinery to regulate the autophagosome closure upon nutrient deprivation. Using high-resolution microscopy, 3D-electron tomography, and protease protection assay, we showed that unclosed autophagosomes accumulated in free1 mutants. Proteomic, cellular and biochemical analysis revealed the mechanistic connection between FREE1 and the ATG conjugation system/ESCRT-III complex in regulating autophagosome closure. Mass spectrometry analysis showed that the evolutionary conserved plant energy sensor SnRK1α1 phosphorylates FREE1 and recruits it to the autophagosomes to promote closure. Mutagenesis of the phosphorylation site on FREE1 caused the autophagosome closure failure. Our findings unveil how cellular energy sensing pathways regulate autophagosome closure to maintain cellular homeostasis.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Autophagosomes , Vesicular Transport Proteins , Arabidopsis/growth & development , Arabidopsis/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism
3.
Curr Neuropharmacol ; 21(3): 482-490, 2023.
Article in English | MEDLINE | ID: mdl-36722478

ABSTRACT

BACKGROUND: The mutated VAPBP56S (vesicle B associated membrane protein - P56S) protein has been described in a Brazilian family and classified as Amyotrophic Lateral Sclerosis type 8 (ALS8). OBJECTIVE: We aimed to study altered biochemical and immunological parameters in cells from ALS8 patients to identify possible biomarkers or therapeutic targets. METHODS: Wild-type VAPB, VAPBP56S, mTOR, proinflammatory cytokines, and oxidant/reducing levels in serum, leucocytes, and cellular lysate from ALS8 patients and health Controls were performed by ELISA, fluorimetry, and spectrophotometry. RESULTS: Our results showed similar levels of mutant and wild-type VAPB in serum and intracellular lysate (p > 0.05) when ALS8 patients and Controls were compared. IL-1ß, IL-6, and IL-18 levels in patients and Controls showed no difference, suggesting an absence of peripheral inflammation (p > 0.05). Oxidative metabolic response, assessed by mitochondrial ROS production, and reductive response by MTT reduction, were higher in the ALS8 group compared to Controls (p < 0.05), although not characterizing typical oxidative stress in ALS8 patients. Total mTOR levels (phosphorylated or non-phosphorylated) of ALS8 patients were significantly lower in serum and higher in intracellular lysate than the mean equivalents in Controls (p < 0.05). A similar result was observed when we quantified the phosphorylated protein (p < 0.05). CONCLUSION: We demonstrate the possibility of using these biochemical and immunological parameters as potential therapeutic targets or biomarkers. Furthermore, by hypothesis, we suggest a hormetic response in which both VAPB forms could coexist in different proportions throughout life. The mutated VAPBP56S production would increase with aging and predominate over the wild-type VAPB levels, determining the onset of symptoms and aggravating the disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Vesicular Transport Proteins , Humans , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Membrane Proteins/genetics , Leukocytes/metabolism , Mutation , TOR Serine-Threonine Kinases
4.
J Cell Biol ; 221(5)2022 06 06.
Article in English | MEDLINE | ID: mdl-35357422

ABSTRACT

VPS13 proteins are proposed to function at contact sites between organelles as bridges for lipids to move directionally and in bulk between organellar membranes. VPS13s are anchored between membranes via interactions with receptors, including both peripheral and integral membrane proteins. Here we present the crystal structure of VPS13s adaptor binding domain (VAB) complexed with a Pro-X-Pro peptide recognition motif present in one such receptor, the integral membrane protein Mcp1p, and show biochemically that other Pro-X-Pro motifs bind the VAB in the same site. We further demonstrate that Mcp1p and another integral membrane protein that interacts directly with human VPS13A, XK, are scramblases. This finding supports an emerging paradigm of a partnership between bulk lipid transport proteins and scramblases. Scramblases can re-equilibrate lipids between membrane leaflets as lipids are removed from or inserted into the cytosolic leaflet of donor and acceptor organelles, respectively, in the course of protein-mediated transport.


Subject(s)
Lipids , Mitochondrial Membranes , Vesicular Transport Proteins , Amino Acid Motifs , Biological Transport , Humans , Lipids/chemistry , Mitochondrial Membranes/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
5.
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
6.
Biochimie ; 192: 72-82, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34634369

ABSTRACT

The transmembrane emp24 domain-containing (TMED) proteins, also called p24 proteins, are members of a family of sorting receptors present in all representatives of the Eukarya and abundantly present in all subcompartments of the early secretory pathway, namely the endoplasmic reticulum (ER), the Golgi, and the intermediate compartment. Although essential during the bidirectional transport between the ER and the Golgi, there is still a lack of information regarding the TMED's structure across different subfamilies. Besides, although the presence of a TMED homo-oligomerization was suggested previously based on crystallographic contacts observed for the isolated Golgi Dynamics (GOLD) domain, no further analyses of its presence in solution were done. Here, we describe the first high-resolution structure of a TMED1 GOLD representative and its biophysical characterization in solution. The crystal structure showed a dimer formation that is also present in solution in a salt-dependent manner, suggesting that the GOLD domain can form homodimers in solution even in the absence of the TMED1 coiled-coil region. A molecular dynamics description of the dimer stabilization, with a phylogenetic analysis of the residues important for the oligomerization and a model for the orientation towards the lipid membrane, are also presented.


Subject(s)
Golgi Apparatus/chemistry , Molecular Docking Simulation , Phylogeny , Vesicular Transport Proteins/chemistry , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Protein Domains , Thermodynamics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
Biomol NMR Assign ; 16(1): 27-30, 2022 04.
Article in English | MEDLINE | ID: mdl-34739631

ABSTRACT

Phafin2 is a peripheral protein that triggers cellular signaling from endosomal and lysosomal compartments. The specific subcellular localization of Phafin2 is mediated by the presence of a tandem of phosphatidylinositol 3-phosphate (PtdIns3P)-binding domains, the pleckstrin homology (PH) and the Fab-1, YOTB, Vac1, and EEA1 (FYVE) domains. The requirement for both domains for binding to PtdIns3P still remains unclear. To understand the molecular interactions of the Phafin2 PH domain in detail, we report its nearly complete 1H, 15N, and 13C backbone resonance assignments.


Subject(s)
Pleckstrin Homology Domains , Vesicular Transport Proteins , Endosomes/metabolism , Endosomes/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
8.
Proteins ; 90(2): 340-350, 2022 02.
Article in English | MEDLINE | ID: mdl-34431571

ABSTRACT

Outbreaks of the Ebola virus (EBOV) continue to occur and while a vaccine and treatment are now available, there remains a dearth of options for those who become sick with EBOV disease. An understanding at the atomic and molecular level of the various steps in the EBOV replication cycle can provide molecular targets for disrupting the virus. An important step in the EBOV replication cycle is the transport of EBOV structural matrix VP40 protein molecules to the plasma membrane inner leaflet, which involves VP40 binding to the host cell's Sec24c protein. Though some VP40 residues involved in the binding are known, the molecular details of VP40-Sec24c binding are not known. We use various molecular computational techniques to investigate the molecular details of how EBOV VP40 binds with the Sec24c complex of the ESCRT-I pathway. We employed different docking programs to identify the VP40-binding site on Sec24c and then performed molecular dynamics simulations to determine the atomic details and binding interactions of the complex. We also investigated how the inter-protein interactions of the complex are affected upon mutations of VP40 amino acids in the Sec24c-binding region. Our results provide a molecular basis for understanding previous coimmunoprecipitation experimental studies. In addition, we found that VP40 can bind to a site on Sec24c that can also bind Sec23 and suggests that VP40 may use the COPII transport mechanism in a manner that may not need the Sec23 protein in order for VP40 to be transported to the plasma membrane.


Subject(s)
Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/virology , Vesicular Transport Proteins , Viral Matrix Proteins , Humans , Protein Binding , Protein Transport , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism
9.
Cells ; 10(12)2021 12 07.
Article in English | MEDLINE | ID: mdl-34943955

ABSTRACT

Commander complex is a 16-protein complex that plays multiple roles in various intracellular events in endosomal cargo and in the regulation of cell homeostasis, cell cycle and immune response. It consists of COMMD1-10, CCDC22, CCDC93, DENND10, VPS26C, VPS29, and VPS35L. These proteins are expressed ubiquitously in the human body, and they have been linked to diseases including Wilson's disease, atherosclerosis, and several types of cancer. In this review we describe the function of the commander complex in endosomal cargo and summarize the individual known roles of COMMD proteins in cell signaling and cancer. It becomes evident that commander complex might be a much more important player in intracellular regulation than we currently understand, and more systematic research on the role of commander complex is required.


Subject(s)
Endosomes/genetics , Multiprotein Complexes/genetics , Protein Transport/genetics , Vesicular Transport Proteins/genetics , Endosomes/ultrastructure , Humans , Intracellular Signaling Peptides and Proteins/genetics , Multiprotein Complexes/ultrastructure , Neoplasms/genetics , Proteins/genetics , Signal Transduction/genetics , Vesicular Transport Proteins/chemistry
10.
Cell Rep ; 37(13): 110182, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965419

ABSTRACT

Whether and how the pathogenic disruptions in endosomal trafficking observed in Alzheimer's disease (AD) are linked to its anatomical vulnerability remain unknown. Here, we began addressing these questions by showing that neurons are enriched with a second retromer core, organized around VPS26b, differentially dedicated to endosomal recycling. Next, by imaging mouse models, we show that the trans-entorhinal cortex, a region most vulnerable to AD, is most susceptible to VPS26b depletion-a finding validated by electrophysiology, immunocytochemistry, and behavior. VPS26b was then found enriched in the trans-entorhinal cortex of human brains, where both VPS26b and the retromer-related receptor SORL1 were found deficient in AD. Finally, by regulating glutamate receptor and SORL1 recycling, we show that VPS26b can mediate regionally selective synaptic dysfunction and SORL1 deficiency. Together with the trans-entorhinal's unique network properties, hypothesized to impose a heavy demand on endosomal recycling, these results suggest a general mechanism that can explain AD's regional vulnerability.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Endosomes/pathology , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/physiology , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Case-Control Studies , Endosomes/metabolism , Female , Humans , LDL-Receptor Related Proteins/genetics , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neuroimaging , Protein Transport , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
11.
Cell Rep ; 37(13): 110174, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965422

ABSTRACT

Lipogenesis plays a critical role in colorectal carcinogenesis, but precisely how remains unclear. Here, we show that ERK2 phosphorylates ME1 at T103, thereby inhibiting its polyubiquitination and proteasomal degradation and enhancing its interaction with USP19. USP19 antagonizes RNF1-mediated ME1 degradation by deubiquitination, which in turn promotes lipid metabolism and NADPH production and suppresses ROS. Meanwhile, ROS dramatically increases PD-L1 mRNA levels through accelerating expression of the transcription factor NRF2. Increased lipid metabolism is correlated with ERK2 activity and colorectal carcinogenesis in human patients. Therefore, the combination of ERK2 inhibitor and anti-PD-L1 antibody significantly inhibits spontaneous and chemically induced colorectal carcinogenesis. Collectively, the USP19-ME1 axis plays a vital role in colorectal carcinogenesis and may also provide a potential therapeutic target.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Colorectal Neoplasms/pathology , Endopeptidases/metabolism , Lipogenesis , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Ubiquitination , Vesicular Transport Proteins/chemistry , Aged , Aged, 80 and over , Aminopyridines/pharmacology , Animals , Carcinogenesis , Case-Control Studies , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Endopeptidases/genetics , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Male , Mice, Inbred C57BL , Middle Aged , Phosphorylation , Pyrroles/pharmacology , Reactive Oxygen Species/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
12.
Int J Mol Sci ; 22(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34948065

ABSTRACT

VAPB (Vesicle-Associated-membrane Protein-associated protein B) is a tail-anchored membrane protein of the endoplasmic reticulum that can also be detected at the inner nuclear membrane. As a component of many contact sites between the endoplasmic reticulum and other organelles, VAPB is engaged in multiple protein interactions with a plethora of binding partners. A mutant version of VAPB, P56S-VAPB, which results from a single point mutation, is involved in a familial form of amyotrophic lateral sclerosis (ALS8). We performed RAPIDS (rapamycin- and APEX-dependent identification of proteins by SILAC) to identify proteins that interact with or are in close proximity to P56S-VAPB. The mutation abrogates the interaction of VAPB with many known binding partners. Here, we identify Sequestosome 1 (SQSTM1), a well-known autophagic adapter protein, as a major interaction/proximity partner of P56S-VAPB. Remarkably, not only the mutant protein, but also wild-type VAPB interacts with SQSTM1, as shown by proximity ligation assays and co-immunoprecipiation experiments.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Point Mutation , Sequestosome-1 Protein/metabolism , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Models, Molecular , Nuclear Envelope/metabolism , Protein Conformation , Protein Transport , Proteomics , Sequestosome-1 Protein/chemistry , Sirolimus/pharmacology , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
13.
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
14.
Biochem Biophys Res Commun ; 582: 86-92, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34695755

ABSTRACT

The cellular glucose level has to be tightly regulated by a variety of cellular processes. One of them is the degradation of gluconeogenic enzymes such as Fbp1, Icl1, Mdh2, and Pck1 by GID (glucose-induced degradation deficient) E3 ubiquitin ligase. The Gid4 component of the GID ligase complex is responsible for recognizing the N-terminal proline residue of the target substrates under normal conditions. However, an alternative N-recognin Gid10 controls the degradation process under stressed conditions. Although Gid10 shares a high sequence similarity with Gid4, their substrate specificities are quite different. Here, we report the structure of Gid10 from Saccharomyces cerevisiae in complex with Pro/N-degron, Pro-Tyr-Ile-Thr, which is almost identical to the sequence of the natural substrate Art2. Although Gid10 shares many structural features with the Gid4 protein from yeast and humans, the current structure explains the unique structural difference for the preference of bulky hydrophobic residue at the second position of Pro/N-degron. Therefore, this study provides a fundamental basis for understanding of the structural diversity and substrate specificity of recognition components in the GID E3 ligase complex involved in the Pro/N-degron pathway.


Subject(s)
Oligopeptides/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Ubiquitin-Protein Ligases/chemistry , Vesicular Transport Proteins/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Models, Molecular , Oligopeptides/metabolism , Proline/chemistry , Proline/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
15.
PLoS Comput Biol ; 17(9): e1008807, 2021 09.
Article in English | MEDLINE | ID: mdl-34555023

ABSTRACT

Early Endosomal Antigen 1 (EEA1) is a key protein in endosomal trafficking and is implicated in both autoimmune and neurological diseases. The C-terminal FYVE domain of EEA1 binds endosomal membranes, which contain phosphatidylinositol-3-phosphate (PI(3)P). Although it is known that FYVE binds PI(3)P specifically, it has not previously been described of how FYVE attaches and binds to endosomal membranes. In this study, we employed both coarse-grained (CG) and atomistic (AT) molecular dynamics (MD) simulations to determine how FYVE binds to PI(3)P-containing membranes. CG-MD showed that the dominant membrane binding mode resembles the crystal structure of EEA1 FYVE domain in complex with inositol-1,3-diphospate (PDB ID 1JOC). FYVE, which is a homodimer, binds the membrane via a hinge mechanism, where the C-terminus of one monomer first attaches to the membrane, followed by the C-terminus of the other monomer. The estimated total binding energy is ~70 kJ/mol, of which 50-60 kJ/mol stems from specific PI(3)P-interactions. By AT-MD, we could partition the binding mode into two types: (i) adhesion by electrostatic FYVE-PI(3)P interaction, and (ii) insertion of amphipathic loops. The AT simulations also demonstrated flexibility within the FYVE homodimer between the C-terminal heads and coiled-coil stem. This leads to a dynamic model whereby the 200 nm long coiled coil attached to the FYVE domain dimer can amplify local hinge-bending motions such that the Rab5-binding domain at the other end of the coiled coil can explore an area of 0.1 µm2 in the search for a second endosome with which to interact.


Subject(s)
Vesicular Transport Proteins/metabolism , Binding Sites , Dimerization , Molecular Dynamics Simulation , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Protein Domains , Static Electricity , Vesicular Transport Proteins/chemistry
16.
Elife ; 102021 08 10.
Article in English | MEDLINE | ID: mdl-34374644

ABSTRACT

Mechanosensitive ion channels mediate transmembrane ion currents activated by mechanical forces. A mechanosensitive ion channel called TACAN was recently reported. We began to study TACAN with the intent to understand how it senses mechanical forces and functions as an ion channel. Using cellular patch-recording methods, we failed to identify mechanosensitive ion channel activity. Using membrane reconstitution methods, we found that TACAN, at high protein concentrations, produces heterogeneous conduction levels that are not mechanosensitive and are most consistent with disruptions of the lipid bilayer. We determined the structure of TACAN using single-particle cryo-electron microscopy and observed that it is a symmetrical dimeric transmembrane protein. Each protomer contains an intracellular-facing cleft with a coenzyme A cofactor, confirmed by mass spectrometry. The TACAN protomer is related in three-dimensional structure to a fatty acid elongase, ELOVL7. Whilst its physiological function remains unclear, we anticipate that TACAN is not a mechanosensitive ion channel.


Subject(s)
Ion Channels/chemistry , Ion Channels/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Animals , Biosensing Techniques , CHO Cells , Cricetulus , Cryoelectron Microscopy , HEK293 Cells , Humans , Ion Channels/genetics , Mechanotransduction, Cellular/physiology , Mice , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism
17.
J Extracell Vesicles ; 10(10): e12132, 2021 08.
Article in English | MEDLINE | ID: mdl-34429859

ABSTRACT

Extracellular vesicles (EVs) are mediators of intercellular communication under both healthy and pathological conditions, including the induction of pro-metastatic traits, but it is not yet known how and where functional cargoes of EVs are delivered to their targets in host cell compartments. We have described that after endocytosis, EVs reach Rab7+ late endosomes and a fraction of these enter the nucleoplasmic reticulum and transport EV biomaterials to the host cell nucleoplasm. Their entry therein and docking to outer nuclear membrane occur through a tripartite complex formed by the proteins VAP-A, ORP3 and Rab7 (VOR complex). Here, we report that the antifungal compound itraconazole (ICZ), but not its main metabolite hydroxy-ICZ or ketoconazole, disrupts the binding of Rab7 to ORP3-VAP-A complexes, leading to inhibition of EV-mediated pro-metastatic morphological changes including cell migration behaviour of colon cancer cells. With novel, smaller chemical drugs, inhibition of the VOR complex was maintained, although the ICZ moieties responsible for antifungal activity and interference with intracellular cholesterol distribution were removed. Knowing that cancer cells hijack their microenvironment and that EVs derived from them determine the pre-metastatic niche, small-sized inhibitors of nuclear transfer of EV cargo into host cells could find cancer therapeutic applications, particularly in combination with direct targeting of cancer cells.


Subject(s)
Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Fatty Acid-Binding Proteins/metabolism , Itraconazole/pharmacology , Nuclear Envelope/metabolism , Vesicular Transport Proteins/metabolism , rab7 GTP-Binding Proteins/metabolism , Active Transport, Cell Nucleus , Antifungal Agents/pharmacology , Cell Line , Cell Movement/drug effects , Cholestenones/pharmacology , Endocytosis , Endosomes/metabolism , Fatty Acid-Binding Proteins/chemistry , Humans , Ketoconazole/pharmacology , Models, Molecular , Saponins/pharmacology , Vesicular Transport Proteins/chemistry , rab7 GTP-Binding Proteins/chemistry
18.
Int J Mol Sci ; 22(16)2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34445480

ABSTRACT

Multivesicular body (MVB)-mediated endosomal sorting and macroautophagy are the main pathways mediating the transport of cellular components to the vacuole and are essential for maintaining cellular homeostasis. The interplay of these two pathways remains poorly understood in plants. In this study, we show that FYVE DOMAIN PROTEIN REQUIRED FOR ENDOSOMAL SORTING 1 (FREE1), which was previously identified as a plant-specific component of the endosomal sorting complex required for transport (ESCRT), essential for MVB biogenesis and plant growth, can be transported to the vacuole for degradation in response to iron deficiency. The vacuolar transport of ubiquitinated FREE1 protein is mediated by the autophagy pathway. As a consequence, the autophagy deficient mutants, atg5-1 and atg7-2, accumulate more endogenous FREE1 protein and display hypersensitivity to iron deficiency. Furthermore, under iron-deficient growth condition autophagy related genes are upregulated to promote the autophagic degradation of FREE1, thereby possibly relieving the repressive effect of FREE1 on iron absorption. Collectively, our findings demonstrate a unique regulatory mode of protein turnover of the ESCRT machinery through the autophagy pathway to respond to iron deficiency in plants.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/metabolism , Autophagy-Related Proteins/genetics , Iron/metabolism , Vesicular Transport Proteins/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Autophagy , Biological Transport , Endosomal Sorting Complexes Required for Transport , Endosomes/metabolism , Mutation , Proteolysis , Ubiquitination
19.
J Mol Biol ; 433(18): 167145, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34229011

ABSTRACT

Transport Protein Particle complexes (TRAPP) are evolutionarily conserved regulators of membrane trafficking, with this mediated by their guanine nucleotide exchange factor (GEF) activity towards Rab GTPases. In metazoans evidence suggests that two different TRAPP complexes exist, TRAPPII and TRAPPIII. These two complexes share a common core of subunits, with complex specific subunits (TRAPPC9 and TRAPPC10 in TRAPPII and TRAPPC8, TRAPPC11, TRAPPC12, TRAPPC13 in TRAPPIII). TRAPPII and TRAPPIII have distinct specificity for GEF activity towards Rabs, with TRAPPIII acting on Rab1, and TRAPPII acting on Rab1 and Rab11. The molecular basis for how these complex specific subunits alter GEF activity towards Rab GTPases is unknown. Here we have used a combination of biochemical assays, hydrogen deuterium exchange mass spectrometry (HDX-MS) and electron microscopy to examine the regulation of TRAPPII and TRAPPIIII complexes in solution and on membranes. GEF assays revealed that TRAPPIII has GEF activity against Rab1 and Rab43, with no detectable activity against the other 18 Rabs tested. The TRAPPIII complex had significant differences in protein dynamics at the Rab binding site compared to TRAPPII, potentially indicating an important role of accessory subunits in altering the active site of TRAPP complexes. Both the TRAPPII and TRAPPIII complexes had enhanced GEF activity on lipid membranes, with HDX-MS revealing numerous conformational changes that accompany membrane association. HDX-MS also identified a membrane binding site in TRAPPC8. Collectively, our results provide insight into the functions of TRAPP complexes and how they can achieve Rab specificity.


Subject(s)
Cell Membrane/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Mammals/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Binding Sites , Guanine Nucleotide Exchange Factors/genetics , Humans , Mammals/genetics , Protein Conformation , Protein Transport , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics
20.
FEBS Lett ; 595(17): 2248-2256, 2021 09.
Article in English | MEDLINE | ID: mdl-34312846

ABSTRACT

The endoplasmic reticulum transmembrane protein vesicle-associated membrane protein-associated protein (VAP) plays a central role in the formation and function of membrane contact sites (MCS) through its interactions with proteins. The major sperm protein (MSP) domain of VAP binds to a variety of sequences which are referred to as FFAT-like motifs. In this study, we investigated the interactions of eight peptides containing FFAT-like motifs with the VAP-A MSP domain (VAP-AMSP ) by solution NMR. Six of eight peptides are specifically bound to VAP-A. Furthermore, we found that the RNA-dependent RNA polymerase of severe acute respiratory syndrome coronavirus 2 has an FFAT-like motif which specifically binds to VAP-AMSP as well as other FFAT-like motifs. Our results will contribute to the discovery of new VAP interactors.


Subject(s)
Coronavirus RNA-Dependent RNA Polymerase/chemistry , Peptides/chemistry , SARS-CoV-2/enzymology , Vesicular Transport Proteins/chemistry , Amino Acid Motifs , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Humans , Nuclear Magnetic Resonance, Biomolecular , Peptides/metabolism , Protein Binding , SARS-CoV-2/metabolism , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...