Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Nat Commun ; 14(1): 906, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36810735

ABSTRACT

Osteoclasts are giant bone-digesting cells that harbor specialized lysosome-related organelles termed secretory lysosomes (SLs). SLs store cathepsin K and serve as a membrane precursor to the ruffled border, the osteoclast's 'resorptive apparatus'. Yet, the molecular composition and spatiotemporal organization of SLs remains incompletely understood. Here, using organelle-resolution proteomics, we identify member a2 of the solute carrier 37 family (Slc37a2) as a SL sugar transporter. We demonstrate in mice that Slc37a2 localizes to the SL limiting membrane and that these organelles adopt a hitherto unnoticed but dynamic tubular network in living osteoclasts that is required for bone digestion. Accordingly, mice lacking Slc37a2 accrue high bone mass owing to uncoupled bone metabolism and disturbances in SL export of monosaccharide sugars, a prerequisite for SL delivery to the bone-lining osteoclast plasma membrane. Thus, Slc37a2 is a physiological component of the osteoclast's unique secretory organelle and a potential therapeutic target for metabolic bone diseases.


Subject(s)
Bone Resorption , Osteoclasts , Mice , Animals , Osteoclasts/metabolism , Biological Transport , Lysosomes/metabolism , Bone and Bones/metabolism , Cell Membrane/metabolism , Bone Resorption/metabolism
2.
Cells ; 11(21)2022 10 25.
Article in English | MEDLINE | ID: mdl-36359754

ABSTRACT

Mammalian retromer complex contributes to multiple early endosome-associated trafficking pathways whose origins are dependent on which sorting nexin (SNX) they are complexed with. In an attempt to dissect out the contribution of individual retromer-SNX complexes, we examined the trafficking of EGFR in detail within a series of KO cell line models. We demonstrated that the depletion of retromer subunit Vps35 leads to decreased EGFR protein levels in resting cells with enhanced association of EGFR with lysosomal compartments. Compared to control cells, the addition of EGF to Vps35 KO cells resulted in a reduced rate of EGFR degradation; AKT activation and cell prolferation rates were elevated, while ERK activation remained relatively unchanged. These observations are consistent with a prolonged temporal association of EGFR within early endosomes due to the inefficiency of early endosome-associated protein trafficking pathways or organelle maturation due to retromer absence. We did not fully delineate the discrete contributions from retromer-associated SNXs to the phenotypes observed from retromer Vps35 depletion. While each of the knock-outs of SNX1/2, SNX3, or SNX27 promotes the enhanced association of EGFR with early endosomal compartments, only the decreased EGF-mediated EGFR degradation was observed in SNX1/2 dKO cells, while the enhanced AKT activation was only increased in SNX3 KO or SNX27 KO cells. Despite this, each of the knock-outs showed increased EGF-stimulated cell proliferation rates.


Subject(s)
Endosomes , Epidermal Growth Factor , Animals , Endosomes/metabolism , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sorting Nexins/genetics , ErbB Receptors/metabolism , Mammals/metabolism
3.
Sci Adv ; 7(49): eabg4007, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34851660

ABSTRACT

The retromer complex (Vps35-Vps26-Vps29) is essential for endosomal membrane trafficking and signaling. Mutation of the retromer subunit Vps35 causes late-onset Parkinson's disease, while viral and bacterial pathogens can hijack the complex during cellular infection. To modulate and probe its function, we have created a novel series of macrocyclic peptides that bind retromer with high affinity and specificity. Crystal structures show that most of the cyclic peptides bind to Vps29 via a Pro-Leu­containing sequence, structurally mimicking known interactors such as TBC1D5 and blocking their interaction with retromer in vitro and in cells. By contrast, macrocyclic peptide RT-L4 binds retromer at the Vps35-Vps26 interface and is a more effective molecular chaperone than reported small molecules, suggesting a new therapeutic avenue for targeting retromer. Last, tagged peptides can be used to probe the cellular localization of retromer and its functional interactions in cells, providing novel tools for studying retromer function.

4.
Essays Biochem ; 65(7): 987-998, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34528672

ABSTRACT

To date, mechanistic treatments targeting the initial cause of Parkinson's disease (PD) are limited due to the underlying biological cause(s) been unclear. Endosomes and their associated cellular homeostasis processes have emerged to have a significant role in the pathophysiology associated with PD. Several variants within retromer complex have been identified and characterised within familial PD patients. The retromer complex represents a key sorting platform within the endosomal system that regulates cargo sorting that maintains cellular homeostasis. In this review, we summarise the current understandings of how PD-associated retromer variants disrupt cellular trafficking and how the retromer complex can interact with other PD-associated genes to contribute to the disease progression.


Subject(s)
Parkinson Disease , Endosomes/genetics , Endosomes/metabolism , Homeostasis , Humans , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Transport
5.
Traffic ; 22(4): 123-136, 2021 04.
Article in English | MEDLINE | ID: mdl-33347683

ABSTRACT

Retromer core complex is an endosomal scaffold that plays a critical role in orchestrating protein trafficking within the endosomal system. Here we characterized the effect of the Parkinson's disease-linked Vps35 D620N in the endo-lysosomal system using Vps35 D620N rescue cell models. Vps35 D620N fully rescues the lysosomal and autophagy defects caused by retromer knock-out. Analogous to Vps35 knock out cells, the endosome-to-trans-Golgi network transport of cation-independent mannose 6-phosphate receptor (CI-M6PR) is impaired in Vps35 D620N rescue cells because of a reduced capacity to form endosome transport carriers. Cells expressing the Vps35 D620N variant have altered endosomal morphology, resulting in smaller, rounder structures with less tubule-like branches. At the molecular level retromer incorporating Vps35 D620N variant has a decreased binding to retromer associated proteins wiskott-aldrich syndrome protein and SCAR homologue (WASH) and SNX3 which are known to associate with retromer to form the endosome transport carriers. Hence, the partial defects on retrograde protein trafficking carriers in the presence of Vps35 D620N represents an altered cellular state able to cause Parkinson's disease.


Subject(s)
Parkinson Disease , Endosomes/metabolism , Humans , Lysosomes/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Protein Transport , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Autophagy ; 17(9): 2200-2216, 2021 09.
Article in English | MEDLINE | ID: mdl-32897127

ABSTRACT

CAV1 (caveolin 1) expression and secretion is associated with prostate cancer (PCa) disease progression, but the mechanisms underpinning CAV1 release remain poorly understood. Numerous studies have shown CAV1 can be secreted within exosome-like vesicles, but antibody-mediated neutralization can mitigate PCa progression; this is suggestive of an inverted (non-exosomal) CAV1 topology. Here we show that CAV1 can be secreted from specific PCa types in an inverted vesicle-associated form consistent with the features of bioactive CAV1 secretion. Characterization of the isolated vesicles by electron microscopy, single-molecule fluorescence microscopy and proteomics reveals they represent a novel class of exosomes ~40 nm in diameter containing ~50-60 copies of CAV1 and, strikingly, are released via a non-canonical secretory macroautophagy/autophagy pathway. This study provides novel insights into a mechanism whereby CAV1 translocates from a normal plasma membrane distribution to an inverted secreted form implicated in PCa disease progression.Abbreviations: 3-MA: 3-methyladenine; APEX: a modified soybean ascorbate peroxidase; ATG5: autophagy related 5; ATG9A: autophagy related 9A; ATG12: autophagy related 12; BHK: baby hamster kidney; C-exosomes: caveolin-exosomes; CAMKK2/CAMKKß: calckum/calmodulin dependent protein kinase kinase 2; CAV1: caveolin 1; DAB: 3,3'-diaminobenzidine; DAPK: death associated protein kinase; EEA1: early endosome antigen 1; EM: electron microscopy; FCS: fluorescence correlation spectroscopy; GBP: GFP/YFP-binding peptide; GFP: green fluorescent protein; GOLGA2: golgin A2; ILVs: intralumenal vesicles; LC3: microtubule-associated protein 1 light chain 3; MBP: maltose binding protein; MTORC1: mechanistic target of rapamycin kinase complex 1; MVBs: multivesicular bodies; PBS: phosphate-buffered saline; PCa: prostate cancer; PI3K: phosphoinositide 3-kinase; PM: plasma membrane; SFM: serum-free medium; TSG101: tumor susceptibility 101; WCL: whole cell lysates; WT: wild type; YFP: yellow fluorescent protein; ßoG: ß-octylglucoside.


Subject(s)
Caveolin 1 , Exosomes , Prostatic Neoplasms , Autophagy , Caveolin 1/metabolism , Exosomes/metabolism , Humans , Male
7.
Nat Commun ; 10(1): 1528, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30948714

ABSTRACT

Phox homology (PX) domains are membrane interacting domains that bind to phosphatidylinositol phospholipids or phosphoinositides, markers of organelle identity in the endocytic system. Although many PX domains bind the canonical endosome-enriched lipid PtdIns3P, others interact with alternative phosphoinositides, and a precise understanding of how these specificities arise has remained elusive. Here we systematically screen all human PX domains for their phospholipid preferences using liposome binding assays, biolayer interferometry and isothermal titration calorimetry. These analyses define four distinct classes of human PX domains that either bind specifically to PtdIns3P, non-specifically to various di- and tri-phosphorylated phosphoinositides, bind both PtdIns3P and other phosphoinositides, or associate with none of the lipids tested. A comprehensive evaluation of PX domain structures reveals two distinct binding sites that explain these specificities, providing a basis for defining and predicting the functional membrane interactions of the entire PX domain protein family.


Subject(s)
Phosphatidylinositols/chemistry , Binding Sites , Calorimetry , Humans , Interferometry , Models, Molecular , Phosphatidylinositols/metabolism , Protein Domains , Sequence Analysis, Protein , Sorting Nexins/chemistry , Sorting Nexins/metabolism
8.
Cell Biol Int ; 43(11): 1234-1244, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30791178

ABSTRACT

GCC88 is a golgin coiled-coil protein at the trans-Golgi (TGN) that functions as a tethering factor for the endosome-derived retrograde transport vesicles. Here, we demonstrate that GCC88 is required for the endosome-to-TGN retrograde transport of the cation-independent mannose 6-phosphate receptor (CI-M6PR). The knockout of GCC88 perturbs the retrieval of CI-M6PR and decreases its cellular level at the steady state, which causes the improper processing of newly synthesized cathepsin-D, a lysosomal hydrolase dependent on CI-M6PR for its delivery to lysosomes. At the whole cell level, the knockout of GCC88 reduces the lysosomal proteolytic capacity but does not impair of the efficiency of autophagy within these cells.


Subject(s)
Cathepsin D/metabolism , Endosomes/metabolism , Golgi Matrix Proteins/physiology , Lysosomes/metabolism , Receptor, IGF Type 2/metabolism , trans-Golgi Network/metabolism , Autophagy , Golgi Matrix Proteins/genetics , HeLa Cells , Humans , Membrane Proteins/metabolism , Protein Transport
9.
Neurobiol Aging ; 77: 144-153, 2019 05.
Article in English | MEDLINE | ID: mdl-30797171

ABSTRACT

There is in vitro evidence that sorting nexin family member 27 (SNX27), a member of the retromer complex, changes the distribution of the amyloid-beta (Aß) precursor protein (APP) to promote its recycling and thereby prevent the production of Aß, the toxic protein associated with Alzheimer's disease (AD). In this study, we analyzed the phenotype of the familial AD APP/PS mouse strain lacking one copy of the SNX27 gene. The reduction in SNX27 expression had no significant effect on the in vivo accumulation of soluble, total, or plaque-deposited Aß, which is overproduced by the familial APP/PS transgenes. Hippocampal structure and cholinergic basal forebrain neuronal health were also unaffected. Nonetheless, mild positive and negative effects of age and/or genotype on spatial navigation performance were observed in SNX27+/- and SNX27+/-APP/PS mice, respectively. These data suggest that downregulation of SNX27 alone does not have long-term negative consequences on spatial memory, but that cognitive dysfunction in the context of high Aß deposition is exacerbated by the cellular or molecular changes induced by reduced SNX27 function.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Down-Regulation/genetics , Down-Regulation/physiology , Gene Expression , Presenilin-1/genetics , Presenilin-1/metabolism , Sorting Nexins/genetics , Sorting Nexins/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Disease Progression , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration , Sorting Nexins/physiology , Spatial Memory
10.
Bio Protoc ; 9(18): e3362, 2019 Sep 20.
Article in English | MEDLINE | ID: mdl-33654860

ABSTRACT

The enrichment of lysosomes is a useful way to study their structure and function. These dynamic vesicles can be enriched from cell cultures in a variety of ways including immunoprecipitation and fluorescence-activated organelle sorting. These methods are extremely precise but often require the transfection and expression of an affinity or fluorophore-tagged lysosomal membrane protein. A simpler approach uses differential density of subcellular organelles, which are characteristic to a particular type of organelle. Separation of organelles along a density-gradient enables fractionation to enrich for specific organelles (such as lysosomes) in their native state. This protocol outlines an optimized method for enriching lysosomes from HeLa cells with a continuous density-gradient that contains Percoll. Gentle cell lysis and extraction conditions yield dense-fractions that are enriched with functional and intact lysosomes, which can be assayed in downstream analyses. This method is quick (conducted in less than 2 h after harvesting cells), and can be easily scaled and optimized for other cell types.

11.
J Cell Biol ; 218(2): 615-631, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30559172

ABSTRACT

Retromer is a peripheral membrane protein complex that coordinates multiple vesicular trafficking events within the endolysosomal system. Here, we demonstrate that retromer is required for the maintenance of normal lysosomal morphology and function. The knockout of retromer subunit Vps35 causes an ultrastructural alteration in lysosomal structure and aberrant lysosome function, leading to impaired autophagy. At the whole-cell level, knockout of retromer Vps35 subunit reduces lysosomal proteolytic capacity as a consequence of the improper processing of lysosomal hydrolases, which is dependent on the trafficking of the cation-independent mannose 6-phosphate receptor (CI-M6PR). Incorporation of CI-M6PR into endosome transport carriers via a retromer-dependent process is restricted to those tethered by GCC88 but not golgin-97 or golgin-245. Finally, we show that this retromer-dependent retrograde cargo trafficking pathway requires SNX3, but not other retromer-associated cargo binding proteins, such as SNX27 or SNX-BAR proteins. Therefore, retromer does contribute to the retrograde trafficking of CI-M6PR required for maturation of lysosomal hydrolases and lysosomal function.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Receptor, IGF Type 2/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Biological Transport, Active/physiology , Endosomes/genetics , Golgi Matrix Proteins/genetics , Golgi Matrix Proteins/metabolism , HeLa Cells , Humans , Lysosomes/genetics , Multiprotein Complexes/genetics , Receptor, IGF Type 2/genetics , Sorting Nexins/genetics , Sorting Nexins/metabolism
12.
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
13.
J Biol Chem ; 293(18): 6802-6811, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29563155

ABSTRACT

Alanine-, serine-, cysteine-preferring transporter 2 (ASCT2, SLC1A5) is responsible for the uptake of glutamine into cells, a major source of cellular energy and a key regulator of mammalian target of rapamycin (mTOR) activation. Furthermore, ASCT2 expression has been reported in several human cancers, making it a potential target for both diagnostic and therapeutic purposes. Here we identify ASCT2 as a membrane-trafficked cargo molecule, sorted through a direct interaction with the PDZ domain of sorting nexin 27 (SNX27). Using both membrane fractionation and subcellular localization approaches, we demonstrate that the majority of ASCT2 resides at the plasma membrane. This is significantly reduced within CrispR-mediated SNX27 knockout (KO) cell lines, as it is missorted into the lysosomal degradation pathway. The reduction of ASCT2 levels in SNX27 KO cells leads to decreased glutamine uptake, which, in turn, inhibits cellular proliferation. SNX27 KO cells also present impaired activation of the mTOR complex 1 (mTORC1) pathway and enhanced autophagy. Taken together, our data reveal a role for SNX27 in glutamine uptake and amino acid-stimulated mTORC1 activation via modulation of ASCT2 intracellular trafficking.


Subject(s)
Amino Acid Transport System ASC/metabolism , Glutamine/metabolism , Minor Histocompatibility Antigens/metabolism , Sorting Nexins/physiology , Autophagy , Cell Cycle , Cell Proliferation , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockdown Techniques , HeLa Cells , Humans , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , PDZ Domains , Protein Transport/physiology , Signal Transduction , Sorting Nexins/chemistry , Sorting Nexins/genetics , Subcellular Fractions/metabolism
14.
FEBS Lett ; 592(7): 1096-1112, 2018 04.
Article in English | MEDLINE | ID: mdl-29210454

ABSTRACT

The endosomal system is critical for the maintenance of intracellular homeostasis, and defects in this system are often linked to neurological disorders. The retromer complex is a critical coordinator of endosomal dynamics and has functional roles in multiple cellular processes through sorting cargoes from endosomes to the trans-Golgi network (TGN) or to the plasma membrane. Mammalian retromer comprises a core Vps26-Vps35-Vps29 trimer and associates with a range of proteins to generate endosomal tubular-vesicular carriers. Alterations in retromer function or its molecular organization have been a rising risk factor for Parkinson's disease (PD). Although genetic evidence has shown several variants within retromer in late-onset PD cases, the exact molecular machineries by which retromer variants induce the development of PD are still not completely elucidated. In this Review, we will focus on the functional roles of retromer in neuronal health, summarize advances in defining the cellular pathological phenotype caused by retromer deficiency or PD-linked retromer variants and discuss the potential clues of how retromer deregulation may contribute to PD pathogenesis.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Endosomes , Neurons , Parkinson Disease , trans-Golgi Network , Animals , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/genetics , Endosomes/metabolism , Endosomes/pathology , Humans , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , trans-Golgi Network/genetics , trans-Golgi Network/metabolism , trans-Golgi Network/pathology
15.
Article in English | MEDLINE | ID: mdl-28664153

ABSTRACT

To invade epithelial cells, Salmonella enterica serovar Typhimurium (S. Typhimurium) induces macropinocytosis through the action of virulence proteins delivered across the host cell membrane via a type III secretion system. We show that after docking at the plasma membrane S. Typhimurium triggers rapid recruitment of cytosolic SNX18, a SH3-PX-BAR domain sorting nexin protein, to the bacteria-induced membrane ruffles and to the nascent Salmonella-containing vacuole. SNX18 recruitment required the inositol-phosphatase activity of the Salmonella effector SopB and an intact phosphoinositide-binding site within the PX domain of SNX18, but occurred independently of Rho-GTPases Rac1 and Cdc42 activation. SNX18 promotes formation of the SCV from the plasma membrane by acting as a scaffold to recruit Dynamin-2 and N-WASP in a process dependent on the SH3 domain of SNX18. Quantification of bacteria uptake revealed that overexpression of SNX18 increased bacteria internalization, whereas a decrease was detected in cells overexpressing the phosphoinositide-binding mutant R303Q, the ΔSH3 mutant, and in cells where endogenous levels of SNX18 were knocked-down. This study identifies SNX18 as a novel target of SopB and suggests a mechanism where S. Typhimurium engages host factors via local manipulation of phosphoinositide composition at the site of invasion to orchestrate the internalization process.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions/physiology , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sorting Nexins/metabolism , Animals , Benzylamines/pharmacology , Cell Membrane/metabolism , Cell Surface Extensions , Cytosol/metabolism , Dynamin II/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mutation , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Pinocytosis , Quinoxalines/pharmacology , RAW 264.7 Cells , Salmonella Infections/microbiology , Sorting Nexins/genetics , Type III Secretion Systems/metabolism , Vacuoles , Virulence , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
16.
Nat Commun ; 8: 14729, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281536

ABSTRACT

Remarkably little is known about how intracellular pathogens exit the host cell in order to infect new hosts. Pathogenic chlamydiae egress by first rupturing their replicative niche (the inclusion) before rapidly lysing the host cell. Here we apply a laser ablation strategy to specifically disrupt the chlamydial inclusion, thereby uncoupling inclusion rupture from the subsequent cell lysis and allowing us to dissect the molecular events involved in each step. Pharmacological inhibition of host cell calpains inhibits inclusion rupture, but not subsequent cell lysis. Further, we demonstrate that inclusion rupture triggers a rapid necrotic cell death pathway independent of BAK, BAX, RIP1 and caspases. Both processes work sequentially to efficiently liberate the pathogen from the host cytoplasm, promoting secondary infection. These results reconcile the pathogen's known capacity to promote host cell survival and induce cell death.


Subject(s)
Calpain/antagonists & inhibitors , Host-Pathogen Interactions , Laser Therapy , Necrosis/parasitology , CRISPR-Cas Systems , Calpain/genetics , Calpain/metabolism , Cell Death/radiation effects , Chlamydia trachomatis/pathogenicity , Chlamydia trachomatis/physiology , Cysteine Proteinase Inhibitors/pharmacology , Gene Editing , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Video , Necrosis/enzymology , Necrosis/genetics , Time-Lapse Imaging , Red Fluorescent Protein
17.
Elife ; 62017 02 22.
Article in English | MEDLINE | ID: mdl-28226239

ABSTRACT

During infection chlamydial pathogens form an intracellular membrane-bound replicative niche termed the inclusion, which is enriched with bacterial transmembrane proteins called Incs. Incs bind and manipulate host cell proteins to promote inclusion expansion and provide camouflage against innate immune responses. Sorting nexin (SNX) proteins that normally function in endosomal membrane trafficking are a major class of inclusion-associated host proteins, and are recruited by IncE/CT116. Crystal structures of the SNX5 phox-homology (PX) domain in complex with IncE define the precise molecular basis for these interactions. The binding site is unique to SNX5 and related family members SNX6 and SNX32. Intriguingly the site is also conserved in SNX5 homologues throughout evolution, suggesting that IncE captures SNX5-related proteins by mimicking a native host protein interaction. These findings thus provide the first mechanistic insights both into how chlamydial Incs hijack host proteins, and how SNX5-related PX domains function as scaffolds in protein complex assembly.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/physiology , Host-Pathogen Interactions , Sorting Nexins/metabolism , Virulence Factors/metabolism , Bacterial Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Sorting Nexins/chemistry , Virulence Factors/chemistry
18.
Curr Protein Pept Sci ; 18(7): 687-701, 2017.
Article in English | MEDLINE | ID: mdl-26965691

ABSTRACT

The retromer complex is a highly conserved membrane trafficking assembly composed of three proteins - Vps26, Vps29 and Vps35 - that were identified over a decade ago in Saccharomyces cerevisiae (S. cerevisiae). Initially, mammalian retromer was shown to sort transmembrane proteins from the endosome to the trans-Golgi network (TGN), though recent work has identified a critical role for retromer in multiple trafficking pathways, including recycling to the plasma membrane and regulation of cell polarity. In recent years, genetic, cellular, pharmacological and animal model studies have identified retromer and its interacting proteins as being linked to familial forms of neurodegenerative diseases such as Alzheimer's (AD) and Parkinson's (PD). Here, this commentary will summarize recently identified point mutations in retromer linked to PD, and explore the molecular functions of retromer that may be relevant to disease progression.


Subject(s)
Alzheimer Disease/genetics , Mutation , Parkinson Disease/genetics , Vesicular Transport Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Gene Expression , Humans , Neurons/metabolism , Neurons/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism
19.
Article in English | MEDLINE | ID: mdl-27625994

ABSTRACT

The intracellular pathogen Salmonella enterica servovar Typhimurium (S.typhimurium) modulates the host cell's phosphoinositide (PI) metabolism to establish its intracellular replicative niche, the Salmonella-containing vacuole (SCV). Upon invasion, phosphoinositide 3-phosphate (PI(3)P) and other early endosomal markers are rapidly recruited to and remain associated with the SCV throughout its early maturation. While the phosphoinositide 3-phosphatase myotubularin 4 (MTMR4) has an established role in regulating autophagy and cellular PI(3)P-content, two processes associated with the intracellular survival of S. typhimurium, a direct role for MTMR4 in Salmonella biology has not been examined. Here we demonstrate that GFP-tagged MTMR4 is recruited to the SCV and infection of cells depleted of endogenous MTMR4 results in a decrease in viable intracellular Salmonella. This reflects a significant increase in the proportion of SCVs with compromised integrity, which targets the compartment for autophagy and consequent bacterial cell death. These findings highlight the importance of PI(3)P regulation to the integrity of the SCV and reveal a novel role for the myotubularins in bacterial pathogenesis.


Subject(s)
Host-Pathogen Interactions , Microbial Viability , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Salmonella typhimurium/physiology , Vacuoles/microbiology , Autophagy , Cell Line , Epithelial Cells/microbiology , Humans , Phosphatidylinositols/metabolism
20.
Nat Struct Mol Biol ; 23(10): 921-932, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27595347

ABSTRACT

Recycling of internalized receptors from endosomal compartments is essential for the receptors' cell-surface homeostasis. Sorting nexin 27 (SNX27) cooperates with the retromer complex in the recycling of proteins containing type I PSD95-Dlg-ZO1 (PDZ)-binding motifs. Here we define specific acidic amino acid sequences upstream of the PDZ-binding motif required for high-affinity engagement of the human SNX27 PDZ domain. However, a subset of SNX27 ligands, such as the ß2 adrenergic receptor and N-methyl-D-aspartate (NMDA) receptor, lack these sequence determinants. Instead, we identified conserved sites of phosphorylation that substitute for acidic residues and dramatically enhance SNX27 interactions. This newly identified mechanism suggests a likely regulatory switch for PDZ interaction and protein transport by the SNX27-retromer complex. Defining this SNX27 binding code allowed us to classify more than 400 potential SNX27 ligands with broad functional implications in signal transduction, neuronal plasticity and metabolite transport.


Subject(s)
Endosomes/metabolism , Sorting Nexins/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Models, Molecular , Molecular Docking Simulation , PDZ Domains , Phosphorylation , Protein Binding , Protein Interaction Maps , Protein Transport , Receptors, Glutamate/metabolism , Sequence Alignment , Signal Transduction , Sorting Nexins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...