Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Annu Rev Biochem ; 89: 21-43, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32569520

ABSTRACT

My coworkers and I have used animal viruses and their interaction with host cells to investigate cellular processes difficult to study by other means. This approach has allowed us to branch out in many directions, including membrane protein characterization, endocytosis, secretion, protein folding, quality control, and glycobiology. At the same time, our aim has been to employ cell biological approaches to expand the fundamental understanding of animal viruses and their pathogenic lifestyles. We have studied mechanisms of host cell entry and the uncoating of incoming viruses as well as the synthesis, folding, maturation, and intracellular movement of viral proteins and molecular assemblies. I have had the privilege to work in institutions in four different countries. The early years in Finland (the University of Helsinki) were followed by 6 years in Germany (European Molecular Biology Laboratory), 16 years in the United States (Yale School of Medicine), and 16 years in Switzerland (ETH Zurich).


Subject(s)
Calnexin/genetics , Calreticulin/genetics , Host-Pathogen Interactions/genetics , Influenza A virus/genetics , Picornaviridae/genetics , Viral Proteins/genetics , Virology/history , Animals , Calnexin/chemistry , Calnexin/metabolism , Calreticulin/chemistry , Calreticulin/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Influenza A virus/metabolism , Picornaviridae/metabolism , Protein Folding , Semliki forest virus/genetics , Semliki forest virus/metabolism , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Internalization
2.
Annu Rev Cell Dev Biol ; 32: 197-222, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27298089

ABSTRACT

Transport of newly synthesized proteins from the endoplasmic reticulum (ER) to the Golgi complex is highly selective. As a general rule, such transport is limited to soluble and membrane-associated secretory proteins that have reached properly folded and assembled conformations. To secure the efficiency, fidelity, and control of this crucial transport step, cells use a combination of mechanisms. The mechanisms are based on selective retention of proteins in the ER to prevent uptake into transport vesicles, on selective capture of proteins in COPII carrier vesicles, on inclusion of proteins in these vesicles by default as part of fluid and membrane bulk flow, and on selective retrieval of proteins from post-ER compartments by retrograde vesicle transport.


Subject(s)
Secretory Pathway , Animals , COP-Coated Vesicles/metabolism , Endoplasmic Reticulum-Associated Degradation , Humans , Protein Transport , Transport Vesicles/metabolism
3.
Annu Rev Biochem ; 79: 803-33, 2010.
Article in English | MEDLINE | ID: mdl-20196649

ABSTRACT

Although viruses are simple in structure and composition, their interactions with host cells are complex. Merely to gain entry, animal viruses make use of a repertoire of cellular processes that involve hundreds of cellular proteins. Although some viruses have the capacity to penetrate into the cytosol directly through the plasma membrane, most depend on endocytic uptake, vesicular transport through the cytoplasm, and delivery to endosomes and other intracellular organelles. The internalization may involve clathrin-mediated endocytosis (CME), macropinocytosis, caveolar/lipid raft-mediated endocytosis, or a variety of other still poorly characterized mechanisms. This review focuses on the cell biology of virus entry and the different strategies and endocytic mechanisms used by animal viruses.


Subject(s)
Endocytosis , Virus Internalization , Animals , Caveolae/metabolism , Clathrin/metabolism , Membrane Microdomains/metabolism , Phagocytosis , Pinocytosis
4.
Proc Natl Acad Sci U S A ; 113(50): E8069-E8078, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27834731

ABSTRACT

Caveolae are invaginated plasma membrane domains involved in mechanosensing, signaling, endocytosis, and membrane homeostasis. Oligomers of membrane-embedded caveolins and peripherally attached cavins form the caveolar coat whose structure has remained elusive. Here, purified Cavin1 60S complexes were analyzed structurally in solution and after liposome reconstitution by electron cryotomography. Cavin1 adopted a flexible, net-like protein mesh able to form polyhedral lattices on phosphatidylserine-containing vesicles. Mutating the two coiled-coil domains in Cavin1 revealed that they mediate distinct assembly steps during 60S complex formation. The organization of the cavin coat corresponded to a polyhedral nano-net held together by coiled-coil segments. Positive residues around the C-terminal coiled-coil domain were required for membrane binding. Purified caveolin 8S oligomers assumed disc-shaped arrangements of sizes that are consistent with the discs occupying the faces in the caveolar polyhedra. Polygonal caveolar membrane profiles were revealed in tomograms of native caveolae inside cells. We propose a model with a regular dodecahedron as structural basis for the caveolae architecture.


Subject(s)
Caveolae/chemistry , Caveolae/metabolism , Caveolin 1/chemistry , Caveolin 1/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Animals , Caveolae/ultrastructure , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Electron, Transmission , Models, Biological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Domains , RNA-Binding Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
5.
Traffic ; 17(4): 351-68, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26650385

ABSTRACT

Human cytomegalovirus (HCMV) is an important and widespread pathogen in the human population. While infection by this ß-herpesvirus in endothelial, epithelial and dendritic cells depends on endocytosis, its entry into fibroblasts is thought to occur by direct fusion of the viral envelope with the plasma membrane. To characterize individual steps during entry in primary human fibroblasts, we employed quantitative assays as well as electron, fluorescence and live cell microscopy in combination with a variety of inhibitory compounds. Our results showed that while infectious entry was pH- and clathrin-independent, it required multiple, endocytosis-related factors and processes. The virions were found to undergo rapid internalization into large vacuoles containing internalized fluid and endosome markers. The characteristics of the internalization process fulfilled major criteria for macropinocytosis. Moreover, we found that soon after addition to fibroblasts the virus rapidly triggered the formation of circular dorsal ruffles in the host cell followed by the generation of large macropinocytic vacuoles. This distinctive form of macropinocytosis has been observed especially in primary cells but has not previously been reported in response to virus stimulation.


Subject(s)
Cytomegalovirus/physiology , Fibroblasts/virology , Pinocytosis , Virus Internalization , Cells, Cultured , Cytomegalovirus/pathogenicity , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans
6.
PLoS Pathog ; 12(3): e1005508, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27030971

ABSTRACT

Phosphoinositide-3-kinases have been shown to be involved in influenza virus pathogenesis. They are targeted directly by virus proteins and are essential for efficient viral replication in infected lung epithelial cells. However, to date the role of PI3K signaling in influenza infection in vivo has not been thoroughly addressed. Here we show that one of the PI3K subunits, p110γ, is in fact critically required for mediating the host's antiviral response. PI3Kγ deficient animals exhibit a delayed viral clearance and increased morbidity during respiratory infection with influenza virus. We demonstrate that p110γ is required for the generation and maintenance of potent antiviral CD8+ T cell responses through the developmental regulation of pulmonary cross-presenting CD103+ dendritic cells under homeostatic and inflammatory conditions. The defect in lung dendritic cells leads to deficient CD8+ T cell priming, which is associated with higher viral titers and more severe disease course during the infection. We thus identify PI3Kγ as a novel key host protective factor in influenza virus infection and shed light on an unappreciated layer of complexity concerning the role of PI3K signaling in this context.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Lung/virology , Orthomyxoviridae Infections/virology , Phosphatidylinositol 3-Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cross-Priming/immunology , Dendritic Cells/immunology , Epithelial Cells/virology , Lung/immunology , Lymphocyte Activation/immunology , Mice , Virus Replication/physiology
7.
Traffic ; 16(8): 814-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25869659

ABSTRACT

The prototypic poxvirus, vaccinia virus (VACV), occurs in two infectious forms, mature virions (MVs) and extracellular virions (EVs). Both enter HeLa cells by inducing macropinocytic uptake. Using confocal microscopy, live-cell imaging, targeted RNAi screening and perturbants of endosome maturation, we analyzed the properties and maturation pathway of the macropinocytic vacuoles containing VACV MVs in HeLa cells. The vacuoles first acquired markers of early endosomes [Rab5, early endosome antigen 1 and phosphatidylinositol(3)P]. Prior to release of virus cores into the cytoplasm, they contained markers of late endosomes and lysosomes (Rab7a, lysosome-associated membrane protein 1 and sorting nexin 3). RNAi screening of endocytic cell factors emphasized the importance of late compartments for VACV infection. Follow-up perturbation analysis showed that infection required Rab7a and PIKfyve, confirming that VACV is a late-penetrating virus dependent on macropinosome maturation. VACV EV infection was inhibited by depletion of many of the same factors, indicating that both infectious particle forms share the need for late vacuolar conditions for penetration.


Subject(s)
Phagocytosis , Phagosomes/metabolism , Vaccinia virus/pathogenicity , Endosomes/metabolism , Endosomes/virology , HeLa Cells , Humans , Lysosomal-Associated Membrane Protein 1/genetics , Lysosomal-Associated Membrane Protein 1/metabolism , Phagosomes/virology , Sorting Nexins/genetics , Sorting Nexins/metabolism , Vaccinia virus/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
8.
Proc Natl Acad Sci U S A ; 111(12): 4548-53, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24616511

ABSTRACT

Systematic genetic perturbation screening in human cells remains technically challenging. Typically, large libraries of chemically synthesized siRNA oligonucleotides are used, each designed to degrade a specific cellular mRNA via the RNA interference (RNAi) mechanism. Here, we report on data from three genome-wide siRNA screens, conducted to uncover host factors required for infection of human cells by two bacterial and one viral pathogen. We find that the majority of phenotypic effects of siRNAs are unrelated to the intended "on-target" mechanism, defined by full complementarity of the 21-nt siRNA sequence to a target mRNA. Instead, phenotypes are largely dictated by "off-target" effects resulting from partial complementarity of siRNAs to multiple mRNAs via the "seed" region (i.e., nucleotides 2-8), reminiscent of the way specificity is determined for endogenous microRNAs. Quantitative analysis enabled the prediction of seeds that strongly and specifically block infection, independent of the intended on-target effect. This prediction was confirmed experimentally by designing oligos that do not have any on-target sequence match at all, yet can strongly reproduce the predicted phenotypes. Our results suggest that published RNAi screens have primarily, and unintentionally, screened the sequence space of microRNA seeds instead of the intended on-target space of protein-coding genes. This helps to explain why previously published RNAi screens have exhibited relatively little overlap. Our analysis suggests a possible way of identifying "seed reagents" for controlling phenotypes of interest and establishes a general strategy for extracting valuable untapped information from past and future RNAi screens.


Subject(s)
Brucella abortus/drug effects , Bunyaviridae/drug effects , MicroRNAs/genetics , Oligonucleotides/pharmacology , RNA Interference , Salmonella typhimurium/drug effects , Base Sequence , Brucella abortus/genetics , Bunyaviridae/genetics , Genes, Bacterial , HeLa Cells , Humans , RNA, Small Interfering/genetics , Salmonella typhimurium/genetics
9.
EMBO J ; 31(10): 2350-64, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22505029

ABSTRACT

Caveolae are specialized domains present in the plasma membrane (PM) of most mammalian cell types. They function in signalling, membrane regulation, and endocytosis. We found that the Eps-15 homology domain-containing protein 2 (EHD2, an ATPase) associated with the static population of PM caveolae. Recruitment to the PM involved ATP binding, interaction with anionic lipids, and oligomerization into large complexes (60-75S) via interaction of the EH domains with intrinsic NPF/KPF motifs. Hydrolysis of ATP was essential for binding of EHD2 complexes to caveolae. EHD2 was found to undergo dynamic exchange at caveolae, a process that depended on a functional ATPase cycle. Depletion of EHD2 by siRNA or expression of a dominant-negative mutant dramatically increased the fraction of mobile caveolar vesicles coming from the PM. Overexpression of EHD2, in turn, caused confinement of cholera toxin B in caveolae. The confining role of EHD2 relied on its capacity to link caveolae to actin filaments. Thus, EHD2 likely plays a key role in adjusting the balance between PM functions of stationary caveolae and the role of caveolae as vesicular carriers.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Caveolae/metabolism , Cell Membrane/metabolism , Adenosine Triphosphate/metabolism , Carrier Proteins/genetics , Gene Deletion , Gene Expression , Gene Silencing , HeLa Cells , Humans , Protein Binding , Protein Interaction Domains and Motifs
10.
PLoS Pathog ; 10(5): e1004162, 2014 May.
Article in English | MEDLINE | ID: mdl-24874089

ABSTRACT

A two-step, high-throughput RNAi silencing screen was used to identify host cell factors required during human papillomavirus type 16 (HPV16) infection. Analysis of validated hits implicated a cluster of mitotic genes and revealed a previously undetermined mechanism for import of the viral DNA (vDNA) into the nucleus. In interphase cells, viruses were endocytosed, routed to the perinuclear area, and uncoated, but the vDNA failed to be imported into the nucleus. Upon nuclear envelope perforation in interphase cells HPV16 infection occured. During mitosis, the vDNA and L2 associated with host cell chromatin on the metaphase plate. Hence, we propose that HPV16 requires nuclear envelope breakdown during mitosis for access of the vDNA to the nucleoplasm. The results accentuate the value of genes found by RNAi screens for investigation of viral infections. The list of cell functions required during HPV16 infection will, moreover, provide a resource for future virus-host cell interaction studies.


Subject(s)
Human papillomavirus 16 , Mitosis/physiology , Nuclear Envelope/metabolism , Oncogene Proteins, Viral/genetics , RNA Interference , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cells, Cultured , DNA, Viral/genetics , Human papillomavirus 16/genetics , Humans
11.
Proc Natl Acad Sci U S A ; 110(27): 11133-8, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776214

ABSTRACT

Human respiratory syncytial virus is a human pathogen that causes severe infection of the respiratory tract. Current information about the structure of the virus and its interaction with host cells is limited. We carried out an electron cryotomographic characterization of cell culture-grown human respiratory syncytial virus to determine the architecture of the virion. The particles ranged from 100 nm to 1,000 nm in diameter and were spherical, filamentous, or a combination of the two. The filamentous morphology correlated with the presence of a cylindrical matrix protein layer linked to the inner leaflet of the viral envelope and with local ordering of the glycoprotein spikes. Recombinant viruses with only the fusion protein in their envelope showed that these glycoproteins were predominantly in the postfusion conformation, but some were also in the prefusion form. The ribonucleocapsids were left-handed, randomly oriented, and curved inside the virions. In filamentous particles, they were often adjacent to an intermediate layer of protein assigned to M2-1 (an envelope-associated protein known to mediate association of ribonucleocapsids with the matrix protein). Our results indicate important differences in structure between the Paramyxovirinae and Pneumovirinae subfamilies within the Paramyxoviridae, and provide fresh insights into host cell exit of a serious pathogen.


Subject(s)
Respiratory Syncytial Virus, Human/ultrastructure , Cell Line , Cryoelectron Microscopy , Electron Microscope Tomography , Humans , Protein Conformation , Respiratory Syncytial Virus, Human/chemistry , Ribonucleoproteins/chemistry , Ribonucleoproteins/ultrastructure , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/ultrastructure
12.
Traffic ; 14(1): 36-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23046100

ABSTRACT

Cholera toxin enters cells via an unusual pathway that involves trafficking through endosomes to the endoplasmic reticulum (ER). Whether the toxin induces its own pathway or travels along a physiological retrograde route is not known. To study its trafficking, we labeled cholera toxin B (CTB) or endogenous plasma membrane proteins with a small chemical compound, benzylguanine, which covalently reacts with the protein SNAP-tag. Using ER-targeted SNAP-tag as reporter, we found that transport of CTB to the ER depends on dynamin-2 and syntaxin 5. Plasma membrane proteins and a fluid-phase marker added to the medium were also transported to the ER. This flux was not affected by exposing cells to CTB but was inhibited by depleting syntaxin 5 and increased by depleting dynamin-2. As a control for confined intracellular localization of ER-targeted SNAP-tag we used adenovirus-5, which traffics to endosomes and then escapes into the cytosol. The virus did not react with ER-targeted SNAP but with cytosolic SNAP. Together, our results establish a new method (SNAP-trap) to study trafficking of different cargo to the ER and the cytosol and provide evidence for the existence of a constitutive pathway from the cell surface to the ER.


Subject(s)
Cytosol/metabolism , Endocytosis , Endoplasmic Reticulum/metabolism , Animals , Cell Line , Cell Line, Tumor , Cholera Toxin/genetics , Cholera Toxin/metabolism , Dynamin II/metabolism , Endosomes/metabolism , Fluorescent Dyes , Humans , Protein Transport , Qa-SNARE Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
EMBO J ; 30(17): 3481-500, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21878991

ABSTRACT

Being deeply connected to signalling, cell dynamics, growth, regulation, and defence, endocytic processes are linked to almost all aspects of cell life and disease. In this review, we focus on endosomes in the classical endocytic pathway, and on the programme of changes that lead to the formation and maturation of late endosomes/multivesicular bodies. The maturation programme entails a dramatic transformation of these dynamic organelles disconnecting them functionally and spatially from early endosomes and preparing them for their unidirectional role as a feeder pathway to lysosomes.


Subject(s)
Endocytosis/physiology , Endosomes/metabolism , ADP-Ribosylation Factor 1/metabolism , Autophagy/physiology , Coat Protein Complex I/metabolism , Endosomes/ultrastructure , Exosomes/metabolism , GTP Phosphohydrolases/metabolism , Humans , Lysosomes/metabolism , Lysosomes/ultrastructure , rab GTP-Binding Proteins/metabolism
14.
EMBO J ; 30(17): 3647-61, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21792173

ABSTRACT

Vaccinia virus (VACV), the model poxvirus, produces two types of infectious particles: mature virions (MVs) and extracellular virions (EVs). EV particles possess two membranes and therefore require an unusual cellular entry mechanism. By a combination of fluorescence and electron microscopy as well as flow cytometry, we investigated the cellular processes that EVs required to infect HeLa cells. We found that EV particles were endocytosed, and that internalization and infection depended on actin rearrangements, activity of Na(+)/H(+) exchangers, and signalling events typical for the macropinocytic mechanism of endocytosis. To promote their internalization, EVs were capable of actively triggering macropinocytosis. EV infection also required vacuolar acidification, and acid exposure in endocytic vacuoles was needed to disrupt the outer EV membrane. Once exposed, the underlying MV-like particle presumably fused its single membrane with the limiting vacuolar membrane. Release of the viral core into the host cell cytosol allowed for productive infection.


Subject(s)
Pinocytosis , Vaccinia virus/physiology , Virion/physiology , Virus Internalization , Actins/metabolism , HeLa Cells , Humans , Signal Transduction , Sodium-Hydrogen Exchangers/metabolism , Vacuoles/metabolism
15.
J Virol ; 88(22): 13029-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165113

ABSTRACT

UNLABELLED: Influenza A virus (IAV) uses the low pH in late endocytic vacuoles as a cue for penetration by membrane fusion. Here, we analyzed the prefusion reactions that prepare the core for uncoating after it has been delivered to the cytosol. We found that this priming process occurs in two steps that are mediated by the envelope-embedded M2 ion channel. The first weakens the interactions between the matrix protein, M1, and the viral ribonucleoprotein bundle. It involves a conformational change in a linker sequence and the C-terminal domain of M1 after exposure to a pH below 6.5. The second step is triggered by a pH of <6.0 and by the influx of K(+) ions. It causes additional changes in M1 as well as a loss of stability in the viral ribonucleoprotein bundle. Our results indicate that both the switch from Na(+) to K(+) in maturing endosomes and the decreasing pH are needed to prime IAV cores for efficient uncoating and infection of the host cell. IMPORTANCE: The entry of IAV involves several steps, including endocytosis and fusion at late endosomes. Entry also includes disassembly of the viral core, which is composed of the viral ribonucleoproteins and the RNA genome. We have found that the uncoating process of IAV is initiated long before the core is delivered into the cytosol. M2, an ion channel in the viral membrane, is activated when the virus passes through early endosomes. Here, we show that protons entering the virus through M2 cause a conformational change in the matrix protein, M1. This weakens interactions between M1 and the viral ribonucleoproteins. A second change was found to occur when the virus enters late endosomes. The preacidified core is then exposed to a high concentration of K(+), which affects the interactions between the ribonucleoproteins. Thus, when cores are finally delivered to the cytosol, they are already partially destabilized and, therefore, uncoating competent and infectious.


Subject(s)
Endosomes/metabolism , Endosomes/virology , Influenza A virus/drug effects , Influenza A virus/physiology , Potassium/metabolism , Viral Matrix Proteins/metabolism , Virus Uncoating/drug effects , Animals , Humans , Hydrogen-Ion Concentration , Protein Binding/drug effects , Protein Conformation/drug effects
16.
J Virol ; 88(15): 8565-78, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850728

ABSTRACT

UNLABELLED: The Bunyaviridae constitute a large family of enveloped animal viruses, many of which are important emerging pathogens. How bunyaviruses enter and infect mammalian cells remains largely uncharacterized. We used two genome-wide silencing screens with distinct small interfering RNA (siRNA) libraries to investigate host proteins required during infection of human cells by the bunyavirus Uukuniemi virus (UUKV), a late-penetrating virus. Sequence analysis of the libraries revealed that many siRNAs in the screens inhibited infection by silencing not only the intended targets but additional genes in a microRNA (miRNA)-like manner. That the 7-nucleotide seed regions in the siRNAs can cause a perturbation in infection was confirmed by using synthetic miRNAs (miRs). One of the miRs tested, miR-142-3p, was shown to interfere with the intracellular trafficking of incoming viruses by regulating the v-SNARE VAMP3, a strong hit shared by both siRNA screens. Inactivation of VAMP3 by the tetanus toxin led to a block in infection. Using fluorescence-based techniques in fixed and live cells, we found that the viruses enter VAMP3(+) endosomal vesicles 5 min after internalization and that colocalization was maximal 15 min thereafter. At this time, LAMP1 was associated with the VAMP3(+) virus-containing endosomes. In cells depleted of VAMP3, viruses were mainly trapped in LAMP1-negative compartments. Together, our results indicated that UUKV relies on VAMP3 for penetration, providing an indication of added complexity in the trafficking of viruses through the endocytic network. IMPORTANCE: Bunyaviruses represent a growing threat to humans and livestock globally. Unfortunately, relatively little is known about these emerging pathogens. We report here the first human genome-wide siRNA screens for a bunyavirus. The screens resulted in the identification of 562 host cell factors with a potential role in cell entry and virus replication. To demonstrate the robustness of our approach, we confirmed and analyzed the role of the v-SNARE VAMP3 in Uukuniemi virus entry and infection. The information gained lays the basis for future research into the cell biology of bunyavirus infection and new antiviral strategies. In addition, by shedding light on serious caveats in large-scale siRNA screening, our experimental and bioinformatics procedures will be valuable in the comprehensive analysis of past and future high-content screening data.


Subject(s)
Gene Silencing , Host-Pathogen Interactions , RNA, Small Interfering/analysis , Uukuniemi virus/physiology , Vesicle-Associated Membrane Protein 3/metabolism , Virus Internalization , Endosomes/chemistry , Endosomes/virology , Epithelial Cells/virology , Genetic Testing , HeLa Cells , Humans , Lysosomal Membrane Proteins/analysis , RNA, Small Interfering/genetics , Time Factors , Vesicle-Associated Membrane Protein 3/genetics
17.
PLoS Pathog ; 9(4): e1003309, 2013.
Article in English | MEDLINE | ID: mdl-23593008

ABSTRACT

Respiratory Syncytial Virus (RSV) is a highly pathogenic member of the Paramyxoviridae that causes severe respiratory tract infections. Reports in the literature have indicated that to infect cells the incoming viruses either fuse their envelope directly with the plasma membrane or exploit clathrin-mediated endocytosis. To study the entry process in human tissue culture cells (HeLa, A549), we used fluorescence microscopy and developed quantitative, FACS-based assays to follow virus binding to cells, endocytosis, intracellular trafficking, membrane fusion, and infection. A variety of perturbants were employed to characterize the cellular processes involved. We found that immediately after binding to cells RSV activated a signaling cascade involving the EGF receptor, Cdc42, PAK1, and downstream effectors. This led to a series of dramatic actin rearrangements; the cells rounded up, plasma membrane blebs were formed, and there was a significant increase in fluid uptake. If these effects were inhibited using compounds targeting Na⁺/H⁺ exchangers, myosin II, PAK1, and other factors, no infection was observed. The RSV was rapidly and efficiently internalized by an actin-dependent process that had all hallmarks of macropinocytosis. Rather than fusing with the plasma membrane, the viruses thus entered Rab5-positive, fluid-filled macropinosomes, and fused with the membranes of these on the average 50 min after internalization. Rab5 was required for infection. To find an explanation for the endocytosis requirement, which is unusual among paramyxoviruses, we analyzed the fusion protein, F, and could show that, although already cleaved by a furin family protease once, it underwent a second, critical proteolytic cleavage after internalization. This cleavage by a furin-like protease removed a small peptide from the F1 subunits, and made the virus infectious.


Subject(s)
Pinocytosis/physiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Respiratory Syncytial Viruses/pathogenicity , Viral Fusion Proteins/metabolism , Virus Internalization , Actins/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Furin/metabolism , HeLa Cells , Hep G2 Cells , Humans , Membrane Fusion , RNA Interference , RNA, Small Interfering , Respiratory Syncytial Virus Infections/metabolism , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rab5 GTP-Binding Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 109(3): 823-8, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22219362

ABSTRACT

Cullin-3 (Cul3) functions as a scaffolding protein in the Bric-a-brac, Tramtrack, Broad-complex (BTB)-Cul3-Rbx1 ubiquitin E3 ligase complex. Here, we report a previously undescribed role for Cul3 complexes in late endosome (LE) maturation. RNAi-mediated depletion of Cul3 results in a trafficking defect of two cargoes of the endolysosomal pathway, influenza A virus (IAV) and epidermal growth factor receptor (EGFR). IAV is able to reach an acidic endosomal compartment, coinciding with LE/lysosome (LY) markers. However, it remains trapped or the capsid is unable to uncoat after penetration into the cytosol. Similarly, activation and subsequent ubiquitination of EGFR appear normal, whereas downstream EGFR degradation is delayed and its ligand EGF accumulates in LE/LYs. Indeed, Cul3-depleted cells display severe morphological defects in LEs that could account for these trafficking defects; they accumulate acidic LE/LYs, and some cells become highly vacuolated, with enlarged Rab7-positive endosomes. Together, these results suggest a crucial role of Cul3 in regulating late steps in the endolysosomal trafficking pathway.


Subject(s)
Cullin Proteins/metabolism , Endosomes/metabolism , Cell Compartmentation , Cell Line, Tumor , Endosomes/virology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Humans , Influenza A virus/physiology , Lysosomes/metabolism , Proteolysis , RNA, Small Interfering/metabolism , Staining and Labeling , Ubiquitination , Virus Internalization
19.
BMC Genomics ; 15: 1162, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25534632

ABSTRACT

BACKGROUND: Large-scale RNAi screening has become an important technology for identifying genes involved in biological processes of interest. However, the quality of large-scale RNAi screening is often deteriorated by off-targets effects. In order to find statistically significant effector genes for pathogen entry, we systematically analyzed entry pathways in human host cells for eight pathogens using image-based kinome-wide siRNA screens with siRNAs from three vendors. We propose a Parallel Mixed Model (PMM) approach that simultaneously analyzes several non-identical screens performed with the same RNAi libraries. RESULTS: We show that PMM gains statistical power for hit detection due to parallel screening. PMM allows incorporating siRNA weights that can be assigned according to available information on RNAi quality. Moreover, PMM is able to estimate a sharedness score that can be used to focus follow-up efforts on generic or specific gene regulators. By fitting a PMM model to our data, we found several novel hit genes for most of the pathogens studied. CONCLUSIONS: Our results show parallel RNAi screening can improve the results of individual screens. This is currently particularly interesting when large-scale parallel datasets are becoming more and more publicly available. Our comprehensive siRNA dataset provides a public, freely available resource for further statistical and biological analyses in the high-content, high-throughput siRNA screening field.


Subject(s)
Genomics/methods , RNA Interference , RNA, Small Interfering/genetics , Cell Line , Gene Library , Genomics/standards , High-Throughput Screening Assays , Host-Pathogen Interactions/genetics , Humans , ROC Curve , Reproducibility of Results
20.
J Virol ; 87(21): 11504-15, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966408

ABSTRACT

The arenavirus Lassa virus (LASV) causes a severe hemorrhagic fever with high mortality in humans. Antigen-presenting cells, in particular dendritic cells (DCs), are early and preferred targets of LASV, and their productive infection contributes to the virus-induced immunosuppression observed in fatal disease. Here, we characterized the role of the C-type lectin DC-specific ICAM-3-grabbing nonintegrin (DC-SIGN) in LASV entry into primary human DCs using a chimera of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) expressing the LASV glycoprotein (rLCMV-LASVGP). We found that differentiation of human primary monocytes into DCs enhanced virus attachment and entry, concomitant with the upregulation of DC-SIGN. LASV and rLCMV-LASVGP bound to DC-SIGN via mannose sugars located on the N-terminal GP1 subunit of LASVGP. We provide evidence that DC-SIGN serves as an attachment factor for rLCMV-LASVGP in monocyte-derived immature dendritic cells (MDDC) and can accelerate the capture of free virus. However, in contrast to the phlebovirus Uukuniemi virus (UUKV), which uses DC-SIGN as an authentic entry receptor, productive infection with rLCMV-LASVGP was less dependent on DC-SIGN. In contrast to the DC-SIGN-mediated cell entry of UUKV, entry of rLCMV-LASVGP in MDDC was remarkably slow and depended on actin, indicating the use of different endocytotic pathways. In sum, our data reveal that DC-SIGN can facilitate cell entry of LASV in human MDDC but that its role seems distinct from the function as an authentic entry receptor reported for phleboviruses.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendritic Cells/virology , Host-Pathogen Interactions , Lassa virus/physiology , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Virus Internalization , Cells, Cultured , Humans , Lassa virus/genetics , Lymphocytic choriomeningitis virus/genetics , Receptors, Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL