Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Immunity ; 54(1): 132-150.e9, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33271119

ABSTRACT

HLA class I (HLA-I) glycoproteins drive immune responses by presenting antigens to cognate CD8+ T cells. This process is often hijacked by tumors and pathogens for immune evasion. Because options for restoring HLA-I antigen presentation are limited, we aimed to identify druggable HLA-I pathway targets. Using iterative genome-wide screens, we uncovered that the cell surface glycosphingolipid (GSL) repertoire determines effective HLA-I antigen presentation. We show that absence of the protease SPPL3 augmented B3GNT5 enzyme activity, resulting in upregulation of surface neolacto-series GSLs. These GSLs sterically impeded antibody and receptor interactions with HLA-I and diminished CD8+ T cell activation. Furthermore, a disturbed SPPL3-B3GNT5 pathway in glioma correlated with decreased patient survival. We show that the immunomodulatory effect could be reversed through GSL synthesis inhibition using clinically approved drugs. Overall, our study identifies a GSL signature that inhibits immune recognition and represents a potential therapeutic target in cancer, infection, and autoimmunity.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , CD8-Positive T-Lymphocytes/immunology , Glioma/immunology , Glycosphingolipids/metabolism , Glycosyltransferases/metabolism , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Immunotherapy/methods , Antigen Presentation , Aspartic Acid Endopeptidases/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/mortality , Glycosphingolipids/immunology , HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Lymphocyte Activation , Signal Transduction , Survival Analysis , Tumor Escape
3.
Nat Commun ; 11(1): 1128, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111832

ABSTRACT

The sterol-regulatory element binding proteins (SREBP) are central transcriptional regulators of lipid metabolism. Using haploid genetic screens we identify the SREBP Regulating Gene (SPRING/C12ORF49) as a determinant of the SREBP pathway. SPRING is a glycosylated Golgi-resident membrane protein and its ablation in Hap1 cells, Hepa1-6 hepatoma cells, and primary murine hepatocytes reduces SREBP signaling. In mice, Spring deletion is embryonic lethal yet silencing of hepatic Spring expression also attenuates the SREBP response. Mechanistically, attenuated SREBP signaling in SPRINGKO cells results from reduced SREBP cleavage-activating protein (SCAP) and its mislocalization to the Golgi irrespective of the cellular sterol status. Consistent with limited functional SCAP in SPRINGKO cells, reintroducing SCAP restores SREBP-dependent signaling and function. Moreover, in line with the role of SREBP in tumor growth, a wide range of tumor cell lines display dependency on SPRING expression. In conclusion, we identify SPRING as a previously unrecognized modulator of SREBP signaling.


Subject(s)
Cholesterol/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Cell Line , Embryonic Development/genetics , Endoplasmic Reticulum/metabolism , Gene Expression , Golgi Apparatus/metabolism , Haploidy , Hepatocytes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Proteins/genetics
4.
Nat Med ; 25(4): 612-619, 2019 04.
Article in English | MEDLINE | ID: mdl-30833751

ABSTRACT

Cancer cells can evade immune surveillance through the expression of inhibitory ligands that bind their cognate receptors on immune effector cells. Expression of programmed death ligand 1 in tumor microenvironments is a major immune checkpoint for tumor-specific T cell responses as it binds to programmed cell death protein-1 on activated and dysfunctional T cells1. The activity of myeloid cells such as macrophages and neutrophils is likewise regulated by a balance between stimulatory and inhibitory signals. In particular, cell surface expression of the CD47 protein creates a 'don't eat me' signal on tumor cells by binding to SIRPα expressed on myeloid cells2-5. Using a haploid genetic screen, we here identify glutaminyl-peptide cyclotransferase-like protein (QPCTL) as a major component of the CD47-SIRPα checkpoint. Biochemical analysis demonstrates that QPCTL is critical for pyroglutamate formation on CD47 at the SIRPα binding site shortly after biosynthesis. Genetic and pharmacological interference with QPCTL activity enhances antibody-dependent cellular phagocytosis and cellular cytotoxicity of tumor cells. Furthermore, interference with QPCTL expression leads to a major increase in neutrophil-mediated killing of tumor cells in vivo. These data identify QPCTL as a novel target to interfere with the CD47 pathway and thereby augment antibody therapy of cancer.


Subject(s)
Aminoacyltransferases/metabolism , Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Receptors, Immunologic/metabolism , Aminoacyltransferases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Mice, Transgenic , Neoplasms/pathology , Opsonin Proteins/metabolism , Pyrrolidonecarboxylic Acid/metabolism
5.
PLoS Pathog ; 14(8): e1007123, 2018 08.
Article in English | MEDLINE | ID: mdl-30080900

ABSTRACT

Endogenous retroviruses (ERVs), remnants of ancient germline infections, comprise 8% of the human genome. The most recently integrated includes human ERV-K (HERV-K) where several envelope (env) sequences remain intact. Viral pseudotypes decorated with one of those Envs are infectious. Using a recombinant vesicular stomatitis virus encoding HERV-K Env as its sole attachment and fusion protein (VSV-HERVK) we conducted a genome-wide haploid genetic screen to interrogate the host requirements for infection. This screen identified 11 genes involved in heparan sulfate biosynthesis. Genetic inhibition or chemical removal of heparan sulfate and addition of excess soluble heparan sulfate inhibit infection. Direct binding of heparin to soluble HERV-K Env and purified VSV-HERVK defines it as critical for viral attachment. Cell surface bound VSV-HERVK particles are triggered to infect on exposure to acidic pH, whereas acid pH pretreatment of virions blocks infection. Testing of additional endogenous HERV-K env sequences reveals they bind heparin and mediate acid pH triggered fusion. This work reconstructs and defines key steps in the infectious entry pathway of an extinct virus.


Subject(s)
Endogenous Retroviruses/physiology , Heparitin Sulfate/metabolism , Viral Envelope Proteins/metabolism , Viral Tropism/physiology , Virus Internalization , Humans
6.
J Cell Sci ; 131(15)2018 08 03.
Article in English | MEDLINE | ID: mdl-30076240

ABSTRACT

In order to replicate, most pathogens need to enter their target cells. Many viruses enter the host cell through an endocytic pathway and hijack endosomes for their journey towards sites of replication. For delivery of their genome to the host cell cytoplasm and to avoid degradation, viruses have to escape this endosomal compartment without host detection. Viruses have developed complex mechanisms to penetrate the endosomal membrane and have evolved to co-opt several host factors to facilitate endosomal escape. Conversely, there is an extensive variety of cellular mechanisms to counteract or impede viral replication. At the level of cell entry, there are cellular defense mechanisms that recognize endosomal membrane damage caused by virus-induced membrane fusion and pore formation, as well as restriction factors that block these processes. In this Cell Science at a Glance article and accompanying poster, we describe the different mechanisms that viruses have evolved to escape the endosomal compartment, as well as the counteracting cellular protection mechanisms. We provide examples for enveloped and non-enveloped viruses, for which we discuss some unique and unexpected cellular responses to virus-entry-induced membrane damage.


Subject(s)
Endosomes/virology , Animals , Humans , Intracellular Membranes/virology , Virus Internalization , Virus Replication/physiology , Viruses/pathogenicity
7.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29899088

ABSTRACT

Cellular antiviral programs can efficiently inhibit viral infection. These programs are often initiated through signaling cascades induced by secreted proteins, such as type I interferons, interleukin-6 (IL-6), or tumor necrosis factor alpha (TNF-α). In the present study, we generated an arrayed library of 756 human secreted proteins to perform a secretome screen focused on the discovery of novel modulators of viral entry and/or replication. The individual secreted proteins were tested for the capacity to inhibit infection by two replication-competent recombinant vesicular stomatitis viruses (VSVs) with distinct glycoproteins utilizing different entry pathways. Fibroblast growth factor 16 (FGF16) was identified and confirmed as the most prominent novel inhibitor of both VSVs and therefore of viral replication, not entry. Importantly, an antiviral interferon signature was completely absent in FGF16-treated cells. Nevertheless, the antiviral effect of FGF16 is broad, as it was evident on multiple cell types and also on infection by coxsackievirus. In addition, other members of the FGF family also inhibited viral infection. Thus, our unbiased secretome screen revealed a novel protein family capable of inducing a cellular antiviral state. This previously unappreciated role of the FGF family may have implications for the development of new antivirals and the efficacy of oncolytic virus therapy.IMPORTANCE Viruses infect human cells in order to replicate, while human cells aim to resist infection. Several cellular antiviral programs have therefore evolved to resist infection. Knowledge of these programs is essential for the design of antiviral therapeutics in the future. The induction of antiviral programs is often initiated by secreted proteins, such as interferons. We hypothesized that other secreted proteins may also promote resistance to viral infection. Thus, we tested 756 human secreted proteins for the capacity to inhibit two pseudotypes of vesicular stomatitis virus (VSV). In this secretome screen on viral infection, we identified fibroblast growth factor 16 (FGF16) as a novel antiviral against multiple VSV pseudotypes as well as coxsackievirus. Subsequent testing of other FGF family members revealed that FGF signaling generally inhibits viral infection. This finding may lead to the development of new antivirals and may also be applicable for enhancing oncolytic virus therapy.


Subject(s)
Fibroblast Growth Factors/pharmacology , Vesicular stomatitis Indiana virus/physiology , Virus Replication/drug effects , Cell Culture Techniques , Cell Line , Culture Media, Conditioned/metabolism , Gene Library , HEK293 Cells , Hep G2 Cells , Humans , Protein Biosynthesis , Vesicular stomatitis Indiana virus/drug effects , Virus Internalization
8.
Cell Host Microbe ; 23(5): 636-643.e5, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29681460

ABSTRACT

Human type A Enteroviruses (EV-As) cause diseases ranging from hand-foot-and-mouth disease to poliomyelitis-like disease. Although cellular receptors are identified for some EV-As, they remain elusive for the majority of EV-As. We identify the cell surface molecule KREMEN1 as an entry receptor for coxsackievirus A10 (CV-A10). Whereas loss of KREMEN1 renders cells resistant to CV-A10 infection, KREMEN1 overexpression enhances CV-A10 binding to the cell surface and increases susceptibility to infection, indicating that KREMEN1 is a rate-limiting factor for CV-A10 infection. Furthermore, the extracellular domain of KREMEN1 binds CV-A10 and functions as a neutralizing agent during infection. Kremen-deficient mice are resistant to CV-A10-induced lethal paralysis, emphasizing the relevance of Kremen for infection in vivo. KREMEN1 is also essential for infection by a phylogenetic and pathogenic related group of EV-As. Collectively these findings highlight the importance of KREMEN1 for these emerging pathogens and its potential as an antiviral therapeutic target.


Subject(s)
Enterovirus A, Human/metabolism , Enterovirus A, Human/pathogenicity , Enterovirus Infections/metabolism , Membrane Proteins/metabolism , Virus Internalization , Animals , Antigens, Surface , Cell Line , Cell Line, Tumor , Enterovirus/pathogenicity , Enterovirus Infections/immunology , Enterovirus Infections/virology , Female , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , Hand, Foot and Mouth Disease/virology , Humans , Male , Membrane Proteins/genetics , Mice , Mutagenesis , Phylogeny , Protein Domains
9.
Cell Host Microbe ; 22(5): 688-696.e5, 2017 Nov 08.
Article in English | MEDLINE | ID: mdl-29120745

ABSTRACT

Arenaviruses cause fatal hemorrhagic disease in humans. Old World arenavirus glycoproteins (GPs) mainly engage α-dystroglycan as a cell-surface receptor, while New World arenaviruses hijack transferrin receptor. However, the Lujo virus (LUJV) GP does not cluster with New or Old World arenaviruses. Using a recombinant vesicular stomatitis virus containing LUJV GP as its sole attachment and fusion protein (VSV-LUJV), we demonstrate that infection is independent of known arenavirus receptor genes. A genome-wide haploid genetic screen identified the transmembrane protein neuropilin 2 (NRP2) and tetraspanin CD63 as factors for LUJV GP-mediated infection. LUJV GP binds the N-terminal domain of NRP2, while CD63 stimulates pH-activated LUJV GP-mediated membrane fusion. Overexpression of NRP2 or its N-terminal domain enhances VSV-LUJV infection, and cells lacking NRP2 are deficient in wild-type LUJV infection. These findings uncover this distinct set of host cell entry factors in LUJV infection and are attractive focus points for therapeutic intervention.


Subject(s)
Lujo virus/physiology , Neuropilin-2/metabolism , Tetraspanin 30/metabolism , Viral Fusion Proteins/metabolism , Viral Proteins/metabolism , Virus Internalization , Carrier Proteins , Cell Line , Host-Pathogen Interactions/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lujo virus/genetics , Lujo virus/pathogenicity , Protein Interaction Domains and Motifs , Receptors, Cell Surface/metabolism , Receptors, Transferrin , Viral Fusion Proteins/genetics , Viral Proteins/genetics
10.
Arterioscler Thromb Vasc Biol ; 37(11): 2064-2074, 2017 11.
Article in English | MEDLINE | ID: mdl-28882874

ABSTRACT

OBJECTIVE: The cellular demand for cholesterol requires control of its biosynthesis by the mevalonate pathway. Regulation of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), a rate-limiting enzyme in this pathway and the target of statins, is a key control point herein. Accordingly, HMGCR is subject to negative and positive regulation. In particular, the ability of oxysterols and intermediates of the mevalonate pathway to stimulate its proteasomal degradation is an exquisite example of metabolically controlled feedback regulation. To define the genetic determinants that govern this process, we conducted an unbiased haploid mammalian genetic screen. APPROACH AND RESULTS: We generated human haploid cells with mNeon fused to endogenous HMGCR using CRISPR/Cas9 and used these cells to interrogate regulation of HMGCR abundance in live cells. This resulted in identification of known and new regulators of HMGCR, and among the latter, UBXD8 (ubiquitin regulatory X domain-containing protein 8), a gene that has not been previously implicated in this process. We demonstrate that UBXD8 is an essential determinant of metabolically stimulated degradation of HMGCR and of cholesterol biosynthesis in multiple cell types. Accordingly, UBXD8 ablation leads to aberrant cholesterol synthesis due to loss of feedback control. Mechanistically, we show that UBXD8 is necessary for sterol-stimulated dislocation of ubiquitylated HMGCR from the endoplasmic reticulum membrane en route to proteasomal degradation, a function dependent on its UBX domain. CONCLUSIONS: We establish UBXD8 as a previously unrecognized determinant that couples flux across the mevalonate pathway to control of cholesterol synthesis and demonstrate the feasibility of applying mammalian haploid genetics to study metabolic traits.


Subject(s)
Blood Proteins/metabolism , Cholesterol/biosynthesis , Haploidy , Hydroxymethylglutaryl CoA Reductases/metabolism , Membrane Proteins/metabolism , Animals , Blood Proteins/genetics , CRISPR-Cas Systems , Endoplasmic Reticulum/enzymology , Enzyme Stability , Feedback, Physiological , Gene Expression Regulation, Enzymologic , Hep G2 Cells , Hepatocytes/enzymology , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Membrane Proteins/genetics , Mevalonic Acid/metabolism , Microscopy, Confocal , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Proteolysis , Rats , Recombinant Fusion Proteins/metabolism , Transfection , Ubiquitination
11.
Nature ; 546(7657): 307-311, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28562590

ABSTRACT

As key executers of biological functions, the activity and abundance of proteins are subjected to extensive regulation. Deciphering the genetic architecture underlying this regulation is critical for understanding cellular signalling events and responses to environmental cues. Using random mutagenesis in haploid human cells, we apply a sensitive approach to directly couple genomic mutations to protein measurements in individual cells. Here we use this to examine a suite of cellular processes, such as transcriptional induction, regulation of protein abundance and splicing, signalling cascades (mitogen-activated protein kinase (MAPK), G-protein-coupled receptor (GPCR), protein kinase B (AKT), interferon, and Wingless and Int-related protein (WNT) pathways) and epigenetic modifications (histone crotonylation and methylation). This scalable, sequencing-based procedure elucidates the genetic landscapes that control protein states, identifying genes that cause very narrow phenotypic effects and genes that lead to broad phenotypic consequences. The resulting genetic wiring map identifies the E3-ligase substrate adaptor KCTD5 (ref. 1) as a negative regulator of the AKT pathway, a key signalling cascade frequently deregulated in cancer. KCTD5-deficient cells show elevated levels of phospho-AKT at S473 that could not be attributed to effects on canonical pathway components. To reveal the genetic requirements for this phenotype, we iteratively analysed the regulatory network linked to AKT activity in the knockout background. This genetic modifier screen exposes suppressors of the KCTD5 phenotype and mechanistically demonstrates that KCTD5 acts as an off-switch for GPCR signalling by triggering proteolysis of Gßγ heterodimers dissociated from the Gα subunit. Although biological networks have previously been constructed on the basis of gene expression, protein-protein associations, or genetic interaction profiles, we foresee that the approach described here will enable the generation of a comprehensive genetic wiring map for human cells on the basis of quantitative protein states.


Subject(s)
Potassium Channels/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Single-Cell Analysis/methods , Cells, Cultured , Haploidy , Heterotrimeric GTP-Binding Proteins/metabolism , Histones/chemistry , Histones/metabolism , Humans , Interferons/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Phenotype , Phosphorylation/genetics , Potassium Channels/deficiency , Potassium Channels/genetics , Proteolysis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway
12.
Nat Microbiol ; 1: 16025, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-27572444

ABSTRACT

Type 3 secretion systems (T3SSs) of bacterial pathogens translocate bacterial effector proteins that mediate disease into the eukaryotic cytosol. Effectors traverse the plasma membrane through a translocon pore formed by T3SS proteins. In a genome-wide selection, we identified the intermediate filament vimentin as required for infection by the T3SS-dependent pathogen S. flexneri. We found that vimentin is required for efficient T3SS translocation of effectors by S. flexneri and other pathogens that use T3SS, Salmonella enterica serovar Typhimurium and Yersinia pseudotuberculosis. Vimentin and the intestinal epithelial intermediate filament keratin 18 interact with the C-terminus of the Shigella translocon pore protein IpaC. Vimentin and its interaction with IpaC are dispensable for pore formation, but are required for stable docking of S. flexneri to cells; moreover, stable docking triggers effector secretion. These findings establish that stable docking of the bacterium specifically requires intermediate filaments, is a process distinct from pore formation, and is a prerequisite for effector secretion.


Subject(s)
Bacterial Adhesion , Salmonella typhimurium/physiology , Shigella flexneri/physiology , Type III Secretion Systems/metabolism , Vimentin/metabolism , Virulence Factors/metabolism , Yersinia pseudotuberculosis/physiology , Animals , Antigens, Bacterial/metabolism , Cell Line , Host-Pathogen Interactions , Humans , Keratin-18/metabolism , Mice , Protein Binding , Protein Transport
13.
mSphere ; 1(2)2016.
Article in English | MEDLINE | ID: mdl-27303731

ABSTRACT

Filoviruses are the causative agents of an increasing number of disease outbreaks in human populations, including the current unprecedented Ebola virus disease (EVD) outbreak in western Africa. One obstacle to controlling these epidemics is our poor understanding of the host range of filoviruses and their natural reservoirs. Here, we investigated the role of the intracellular filovirus receptor, Niemann-Pick C1 (NPC1) as a molecular determinant of Ebola virus (EBOV) host range at the cellular level. Whereas human cells can be infected by EBOV, a cell line derived from a Russell's viper (Daboia russellii) (VH-2) is resistant to infection in an NPC1-dependent manner. We found that VH-2 cells are resistant to EBOV infection because the Russell's viper NPC1 ortholog bound poorly to the EBOV spike glycoprotein (GP). Analysis of panels of viper-human NPC1 chimeras and point mutants allowed us to identify a single amino acid residue in NPC1, at position 503, that bidirectionally influenced both its binding to EBOV GP and its viral receptor activity in cells. Significantly, this single residue change perturbed neither NPC1's endosomal localization nor its housekeeping role in cellular cholesterol trafficking. Together with other recent work, these findings identify sequences in NPC1 that are important for viral receptor activity by virtue of their direct interaction with EBOV GP and suggest that they may influence filovirus host range in nature. Broader surveys of NPC1 orthologs from vertebrates may delineate additional sequence polymorphisms in this gene that control susceptibility to filovirus infection. IMPORTANCE Identifying cellular factors that determine susceptibility to infection can help us understand how Ebola virus is transmitted. We asked if the EBOV receptor Niemann-Pick C1 (NPC1) could explain why reptiles are resistant to EBOV infection. We demonstrate that cells derived from the Russell's viper are not susceptible to infection because EBOV cannot bind to viper NPC1. This resistance to infection can be mapped to a single amino acid residue in viper NPC1 that renders it unable to bind to EBOV GP. The newly solved structure of EBOV GP bound to NPC1 confirms our findings, revealing that this residue dips into the GP receptor-binding pocket and is therefore critical to the binding interface. Consequently, this otherwise well-conserved residue in vertebrate species influences the ability of reptilian NPC1 proteins to bind to EBOV GP, thereby affecting viral host range in reptilian cells.

14.
Science ; 344(6191): 1506-10, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24970085

ABSTRACT

Lassa virus spreads from a rodent to humans and can lead to lethal hemorrhagic fever. Despite its broad tropism, chicken cells were reported 30 years ago to resist infection. We found that Lassa virus readily engaged its cell-surface receptor α-dystroglycan in avian cells, but virus entry in susceptible species involved a pH-dependent switch to an intracellular receptor, the lysosome-resident protein LAMP1. Iterative haploid screens revealed that the sialyltransferase ST3GAL4 was required for the interaction of the virus glycoprotein with LAMP1. A single glycosylated residue in LAMP1, present in susceptible species but absent in birds, was essential for interaction with the Lassa virus envelope protein and subsequent infection. The resistance of Lamp1-deficient mice to Lassa virus highlights the relevance of this receptor switch in vivo.


Subject(s)
Lassa virus/physiology , Lysosomal-Associated Membrane Protein 1/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Cells, Cultured , Chickens , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Humans , Hydrogen-Ion Concentration , Lassa Fever/virology , Lysosomal-Associated Membrane Protein 1/chemistry , Lysosomes/metabolism , Lysosomes/virology , Mice , Mice, Knockout , Molecular Sequence Data , Protein Binding , Sialyltransferases/metabolism , beta-Galactoside alpha-2,3-Sialyltransferase
15.
Science ; 340(6131): 479-83, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23519211

ABSTRACT

Glycosylated α-dystroglycan (α-DG) serves as cellular entry receptor for multiple pathogens, and defects in its glycosylation cause hereditary Walker-Warburg syndrome (WWS). At least eight proteins are critical to glycosylate α-DG, but many genes mutated in WWS remain unknown. To identify modifiers of α-DG, we performed a haploid screen for Lassa virus entry, a hemorrhagic fever virus causing thousands of deaths annually that hijacks glycosylated α-DG to enter cells. In complementary screens, we profiled cells for absence of α-DG carbohydrate chains or biochemically related glycans. This revealed virus host factors and a suite of glycosylation units, including all known Walker-Warburg genes and five additional factors critical for the modification of α-DG. Our findings accentuate the complexity of this posttranslational feature and point out genes defective in dystroglycanopathies.


Subject(s)
Dystroglycans/metabolism , Host-Pathogen Interactions/genetics , Lassa Fever/genetics , Lassa virus/physiology , Membrane Proteins/genetics , Proteome/metabolism , Virus Internalization , Walker-Warburg Syndrome/genetics , Amino Acid Sequence , Cell Line , Female , Glycosylation , Haploidy , Humans , Infant , Lassa Fever/virology , Male , Molecular Sequence Data , Mutation , Pedigree , Pentosyltransferases
16.
PLoS Pathog ; 8(5): e1002710, 2012.
Article in English | MEDLINE | ID: mdl-22615569

ABSTRACT

Immunological checkpoints, such as the inhibitory CD200 receptor (CD200R), play a dual role in balancing the immune system during microbial infection. On the one hand these inhibitory signals prevent excessive immune mediated pathology but on the other hand they may impair clearance of the pathogen. We studied the influence of the inhibitory CD200-CD200R axis on clearance and pathology in two different virus infection models. We find that lack of CD200R signaling strongly enhances type I interferon (IFN) production and viral clearance and improves the outcome of mouse hepatitis corona virus (MHV) infection, particularly in female mice. MHV clearance is known to be dependent on Toll like receptor 7 (TLR7)-mediated type I IFN production and sex differences in TLR7 responses previously have been reported for humans. We therefore hypothesize that CD200R ligation suppresses TLR7 responses and that release of this inhibition enlarges sex differences in TLR7 signaling. This hypothesis is supported by our findings that in vivo administration of synthetic TLR7 ligand leads to enhanced type I IFN production, particularly in female Cd200(-/-) mice and that CD200R ligation inhibits TLR7 signaling in vitro. In influenza A virus infection we show that viral clearance is determined by sex but not by CD200R signaling. However, absence of CD200R in influenza A virus infection results in enhanced lung neutrophil influx and pathology in females. Thus, CD200-CD200R and sex are host factors that together determine the outcome of viral infection. Our data predict a sex bias in both beneficial and pathological immune responses to virus infection upon therapeutic targeting of CD200-CD200R.


Subject(s)
Antigens, CD/metabolism , Coronavirus Infections/immunology , Influenza A virus/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Orthomyxoviridae Infections/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Animals , Antigens, CD/genetics , Female , Influenza A virus/pathogenicity , Interferon Type I/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Murine hepatitis virus , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Sex Characteristics , Signal Transduction
17.
EMBO J ; 31(8): 1947-60, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22395071

ABSTRACT

Ebola and Marburg filoviruses cause deadly outbreaks of haemorrhagic fever. Despite considerable efforts, no essential cellular receptors for filovirus entry have been identified. We showed previously that Niemann-Pick C1 (NPC1), a lysosomal cholesterol transporter, is required for filovirus entry. Here, we demonstrate that NPC1 is a critical filovirus receptor. Human NPC1 fulfills a cardinal property of viral receptors: it confers susceptibility to filovirus infection when expressed in non-permissive reptilian cells. The second luminal domain of NPC1 binds directly and specifically to the viral glycoprotein, GP, and a synthetic single-pass membrane protein containing this domain has viral receptor activity. Purified NPC1 binds only to a cleaved form of GP that is generated within cells during entry, and only viruses containing cleaved GP can utilize a receptor retargeted to the cell surface. Our findings support a model in which GP cleavage by endosomal cysteine proteases unmasks the binding site for NPC1, and GP-NPC1 engagement within lysosomes promotes a late step in entry proximal to viral escape into the host cytoplasm. NPC1 is the first known viral receptor that recognizes its ligand within an intracellular compartment and not at the plasma membrane.


Subject(s)
Carrier Proteins/metabolism , Ebolavirus/physiology , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Models, Molecular , Niemann-Pick C1 Protein , Protein Binding , Viperidae , Viral Envelope Proteins/chemistry
18.
Virology ; 419(2): 72-83, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21907381

ABSTRACT

Ebola virus (EBOV) has been reported to enter cultured cell lines via a dynamin-2-independent macropinocytic pathway or clathrin-mediated endocytosis. The route(s) of productive EBOV internalization into physiologically relevant cell types remain unexplored, and viral-host requirements for this process are incompletely understood. Here, we use electron microscopy and complementary chemical and genetic approaches to demonstrate that the viral glycoprotein, GP, induces macropinocytic uptake of viral particles into cells. GP's highly-glycosylated mucin domain is dispensable for virus-induced macropinocytosis, arguing that interactions between other sequences in GP and the host cell surface are responsible. Unexpectedly, we also found a requirement for the large GTPase dynamin-2, which is proposed to be dispensable for several types of macropinocytosis. Our results provide evidence that EBOV uses an atypical dynamin-dependent macropinocytosis-like entry pathway to enter Vero cells, adherent human peripheral blood-derived monocytes, and a mouse dendritic cell line.


Subject(s)
Dynamin II/metabolism , Ebolavirus/physiology , Pinocytosis , Viral Envelope Proteins/metabolism , Virus Internalization , Actins/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Dendritic Cells , Dynamin II/antagonists & inhibitors , Dynamin II/genetics , Ebolavirus/metabolism , Humans , Hydrazones/pharmacology , Mice , Microscopy, Electron , Pinocytosis/drug effects , Vero Cells , Vesiculovirus/drug effects , Virus Internalization/drug effects
19.
Nature ; 477(7364): 340-3, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21866103

ABSTRACT

Infections by the Ebola and Marburg filoviruses cause a rapidly fatal haemorrhagic fever in humans for which no approved antivirals are available. Filovirus entry is mediated by the viral spike glycoprotein (GP), which attaches viral particles to the cell surface, delivers them to endosomes and catalyses fusion between viral and endosomal membranes. Additional host factors in the endosomal compartment are probably required for viral membrane fusion; however, despite considerable efforts, these critical host factors have defied molecular identification. Here we describe a genome-wide haploid genetic screen in human cells to identify host factors required for Ebola virus entry. Our screen uncovered 67 mutations disrupting all six members of the homotypic fusion and vacuole protein-sorting (HOPS) multisubunit tethering complex, which is involved in the fusion of endosomes to lysosomes, and 39 independent mutations that disrupt the endo/lysosomal cholesterol transporter protein Niemann-Pick C1 (NPC1). Cells defective for the HOPS complex or NPC1 function, including primary fibroblasts derived from human Niemann-Pick type C1 disease patients, are resistant to infection by Ebola virus and Marburg virus, but remain fully susceptible to a suite of unrelated viruses. We show that membrane fusion mediated by filovirus glycoproteins and viral escape from the vesicular compartment require the NPC1 protein, independent of its known function in cholesterol transport. Our findings uncover unique features of the entry pathway used by filoviruses and indicate potential antiviral strategies to combat these deadly agents.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Ebolavirus/physiology , Membrane Glycoproteins/metabolism , Virus Internalization , Animals , Biological Transport , Carrier Proteins/genetics , Cell Line , Endosomes/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Genome, Human/genetics , Glycoproteins/metabolism , Haploidy , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/metabolism , Host-Pathogen Interactions/genetics , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Marburg Virus Disease/drug therapy , Marburg Virus Disease/metabolism , Marburgvirus/physiology , Membrane Fusion/genetics , Membrane Fusion/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/deficiency , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation/genetics , Niemann-Pick C1 Protein , Niemann-Pick Diseases/pathology , Niemann-Pick Diseases/virology , Receptors, Virus/metabolism , Vesicular Transport Proteins , Viral Fusion Proteins/metabolism
20.
J Virol ; 84(15): 7869-79, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484504

ABSTRACT

The ubiquitin-proteasome system (UPS) is a key player in regulating the intracellular sorting and degradation of proteins. In this study we investigated the role of the UPS in different steps of the coronavirus (CoV) infection cycle. Inhibition of the proteasome by different chemical compounds (i.e., MG132, epoxomicin, and Velcade) appeared to not only impair entry but also RNA synthesis and subsequent protein expression of different CoVs (i.e., mouse hepatitis virus [MHV], feline infectious peritonitis virus, and severe acute respiratory syndrome CoV). MHV assembly and release were, however, not appreciably affected by these compounds. The inhibitory effect on CoV protein expression did not appear to result from a general inhibition of translation due to induction of a cellular stress response by the inhibitors. Stress-induced phosphorylation of eukaryotic translation initiation factor 2alpha (eIF2alpha) generally results in impaired initiation of protein synthesis, but the sensitivity of MHV infection to proteasome inhibitors was unchanged in cells lacking a phosphorylatable eIF2alpha. MHV infection was affected not only by inhibition of the proteasome but also by interfering with protein ubiquitination. Viral protein expression was reduced in cells expressing a temperature-sensitive ubiquitin-activating enzyme E1 at the restrictive temperature, as well as in cells in which ubiquitin was depleted by using small interfering RNAs. Under these conditions, the susceptibility of the cells to virus infection was, however, not affected, excluding an important role of ubiquitination in virus entry. Our observations reveal an important role of the UPS in multiple steps of the CoV infection cycle and identify the UPS as a potential drug target to modulate the impact of CoV infection.


Subject(s)
Coronavirus Infections/virology , Coronavirus, Feline/pathogenicity , Murine hepatitis virus/pathogenicity , Proteasome Endopeptidase Complex/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Ubiquitin/metabolism , Animals , Boronic Acids/pharmacology , Bortezomib , Cats , Cell Line , Chlorocebus aethiops , Leupeptins/pharmacology , Mice , Oligopeptides/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Virus Internalization , Virus Release , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...