Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Cells ; 10(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33652973

ABSTRACT

Coronaviruses (CoVs) assemble by budding into the lumen of the intermediate compartment (IC) at the endoplasmic reticulum (ER)-Golgi interface. However, why CoVs have chosen the IC as their intracellular site of assembly and how progeny viruses are delivered from this compartment to the extracellular space has remained unclear. Here we address these enigmatic late events of the CoV life cycle in light of recently described properties of the IC. Of particular interest are the emerging spatial and functional connections between IC elements and recycling endosomes (REs), defined by the GTPases Rab1 and Rab11, respectively. The establishment of IC-RE links at the cell periphery, around the centrosome and evidently also at the noncompact zones of the Golgi ribbon indicates that-besides traditional ER-Golgi communication-the IC also promotes a secretory process that bypasses the Golgi stacks, but involves its direct connection with the endocytic recycling system. The initial confinement of CoVs to the lumen of IC-derived large transport carriers and their preferential absence from Golgi stacks is consistent with the idea that they exit cells following such an unconventional route. In fact, CoVs may share this pathway with other intracellularly budding viruses, lipoproteins, procollagen, and/or protein aggregates experimentally introduced into the IC lumen.


Subject(s)
Endoplasmic Reticulum/virology , Extracellular Space/virology , Golgi Apparatus/virology , Intracellular Membranes/virology , SARS-CoV-2/physiology , Secretory Pathway , Virus Release , Animals , COVID-19/therapy , COVID-19/virology , Centrosome/metabolism , Extracellular Space/metabolism , Golgi Apparatus/metabolism , Humans , Protein Transport
2.
Plant Physiol ; 180(3): 1375-1388, 2019 07.
Article in English | MEDLINE | ID: mdl-31019004

ABSTRACT

Turnip mosaic virus (TuMV) reorganizes the endomembrane system of the infected cell to generate endoplasmic-reticulum-derived motile vesicles containing viral replication complexes. The membrane-associated viral protein 6K2 plays a key role in the formation of these vesicles. Using confocal microscopy, we observed that this viral protein, a marker for viral replication complexes, localized in the extracellular space of infected Nicotiana benthamiana leaves. Previously, we showed that viral RNA is associated with multivesicular bodies (MVBs). Here, using transmission electron microscopy, we observed the proliferation of MVBs during infection and their fusion with the plasma membrane that resulted in the release of their intraluminal vesicles in the extracellular space. Immunogold labeling with a monoclonal antibody that recognizes double-stranded RNA indicated that the released vesicles contained viral RNA. Focused ion beam-extreme high-resolution scanning electron microscopy was used to generate a three-dimensional image that showed extracellular vesicles in the cell wall. The presence of TuMV proteins in the extracellular space was confirmed by proteomic analysis of purified extracellular vesicles from N benthamiana and Arabidopsis (Arabidopsis thaliana). Host proteins involved in biotic defense and in interorganelle vesicular exchange were also detected. The association of extracellular vesicles with viral proteins and RNA emphasizes the implication of the plant extracellular space in viral infection.


Subject(s)
Extracellular Space/metabolism , Multivesicular Bodies/metabolism , Plant Leaves/metabolism , Potyvirus/metabolism , Arabidopsis/metabolism , Arabidopsis/virology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Extracellular Space/virology , Host-Pathogen Interactions , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Multivesicular Bodies/ultrastructure , Multivesicular Bodies/virology , Plant Leaves/virology , Potyvirus/genetics , Potyvirus/physiology , Proteomics/methods , RNA, Viral/genetics , RNA, Viral/metabolism , Nicotiana/metabolism , Nicotiana/virology , Viral Proteins/metabolism , Virus Replication/genetics
3.
Nat Commun ; 9(1): 5278, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30538244

ABSTRACT

Regulation of intracellular pH is critically important for many cellular functions. The quantification of proton extrusion in different types of cells and physiological conditions is pivotal to fully elucidate the mechanisms of pH homeostasis. Here we show the use of gold nanoparticles (AuNP) to create a high spatial resolution sensor for measuring extracellular pH in proximity of the cell membrane. We test the sensor on HepG2 liver cancer cells and MKN28 gastric cancer cells before and after inhibition of Na+/H+ exchanger. The gold surface conjugation strategy is conceived with a twofold purpose: i) to anchor the AuNP to the membrane proteins and ii) to quantify the local pH from AuNP using surface enhanced Raman spectroscopy (SERS). The nanometer size of the cell membrane anchored sensor and the use of SERS enable us to visualize highly localized variation of pH induced by H+ extrusion, which is particularly upregulated in cancer cells.


Subject(s)
Extracellular Space/chemistry , Extracellular Space/virology , Gold/chemistry , Metal Nanoparticles/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , Extracellular Space/metabolism , Hep G2 Cells , Humans , Hydrogen/metabolism , Hydrogen-Ion Concentration , Sodium/metabolism , Spectrum Analysis, Raman
4.
J Virol ; 90(10): 5163-5175, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26984724

ABSTRACT

UNLABELLED: Intercellular nanotube connections have been identified as an alternative pathway for cellular spreading of certain viruses. In cells infected with porcine reproductive and respiratory syndrome virus (PRRSV), nanotubes were observed connecting two distant cells with contiguous membranes, with the core infectious viral machinery (viral RNA, certain replicases, and certain structural proteins) present in/on the intercellular nanotubes. Live-cell movies tracked the intercellular transport of a recombinant PRRSV that expressed green fluorescent protein (GFP)-tagged nsp2. In MARC-145 cells expressing PRRSV receptors, GFP-nsp2 moved from one cell to another through nanotubes in the presence of virus-neutralizing antibodies. Intercellular transport of viral proteins did not require the PRRSV receptor as it was observed in receptor-negative HEK-293T cells after transfection with an infectious clone of GFP-PRRSV. In addition, GFP-nsp2 was detected in HEK-293T cells cocultured with recombinant PRRSV-infected MARC-145 cells. The intercellular nanotubes contained filamentous actin (F-actin) with myosin-associated motor proteins. The F-actin and myosin IIA were identified as coprecipitates with PRRSV nsp1ß, nsp2, nsp2TF, nsp4, nsp7-nsp8, GP5, and N proteins. Drugs inhibiting actin polymerization or myosin IIA activation prevented nanotube formation and viral clusters in virus-infected cells. These data lead us to propose that PRRSV utilizes the host cell cytoskeletal machinery inside nanotubes for efficient cell-to-cell spread. This form of virus transport represents an alternative pathway for virus spread, which is resistant to the host humoral immune response. IMPORTANCE: Extracellular virus particles transmit infection between organisms, but within infected hosts intercellular infection can be spread by additional mechanisms. In this study, we describe an alternative pathway for intercellular transmission of PRRSV in which the virus uses nanotube connections to transport infectious viral RNA, certain replicases, and certain structural proteins to neighboring cells. This process involves interaction of viral proteins with cytoskeletal proteins that form the nanotube connections. Intercellular viral spread through nanotubes allows the virus to escape the neutralizing antibody response and may contribute to the pathogenesis of viral infections. The development of strategies that interfere with this process could be critical in preventing the spread of viral infection.


Subject(s)
Extracellular Space/virology , Intercellular Junctions/virology , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Cytoskeletal Proteins/metabolism , Extracellular Space/physiology , Green Fluorescent Proteins , HEK293 Cells , Host-Pathogen Interactions , Humans , Nanotubes , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , RNA, Viral , Swine , Transfection , Viral Proteins/metabolism , Virion/physiology
5.
J Virol ; 89(14): 7038-52, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25926655

ABSTRACT

UNLABELLED: The entry of human papillomaviruses into host cells is a complex process. It involves conformational changes at the cell surface, receptor switching, internalization by a novel endocytic mechanism, uncoating in endosomes, trafficking of a subviral complex to the Golgi complex, and nuclear entry during mitosis. Here, we addressed how the stabilizing contacts in the capsid of human papillomavirus 16 (HPV16) may be reversed to allow uncoating of the viral genome. Using biochemical and cell-biological analyses, we determined that the major capsid protein L1 underwent proteolytic cleavage during entry. In addition to a dispensable cathepsin-mediated proteolysis that occurred likely after removal of capsomers from the subviral complex in endosomes, at least two further proteolytic cleavages of L1 were observed, one of which was independent of the low-pH environment of endosomes. This cleavage occurred extracellularly. Further analysis showed that the responsible protease was the secreted trypsin-like serine protease kallikrein-8 (KLK8) involved in epidermal homeostasis and wound healing. Required for infection, the cleavage was facilitated by prior interaction of viral particles with heparan sulfate proteoglycans. KLK8-mediated cleavage was crucial for further conformational changes exposing an important epitope of the minor capsid protein L2. Occurring independently of cyclophilins and of furin that mediate L2 exposure, KLK8-mediated cleavage of L1 likely facilitated access to L2, located in the capsid lumen, and potentially uncoating. Since HPV6 and HPV18 also required KLK8 for entry, we propose that the KLK8-dependent entry step is conserved. IMPORTANCE: Our analysis of the proteolytic processing of incoming HPV16, an etiological agent of cervical cancer, demonstrated that the capsid is cleaved extracellularly by a serine protease active during wound healing and that this cleavage was crucial for infection. The cleavage of L1 is one of at least four structural alterations that prime the virus extracellularly for receptor switching, internalization, and possibly uncoating. This step was also important for HPV6 and HPV18, which may suggest that it is conserved among the papillomaviruses. This study advances the understanding of how HPV16 initially infects cells, strengthens the notion that wounding facilitates infection of epidermal tissue, and may help the development of antiviral measures.


Subject(s)
Capsid Proteins/metabolism , Extracellular Space/enzymology , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Kallikreins/metabolism , Oncogene Proteins, Viral/metabolism , Protein Processing, Post-Translational , Virus Internalization , Extracellular Space/virology , HeLa Cells , Humans , Proteolysis
6.
J Antimicrob Chemother ; 70(3): 750-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25433008

ABSTRACT

OBJECTIVES: Cenicriviroc is a potent antagonist of the chemokine coreceptors 5 and 2 (CCR5/CCR2) and blocks HIV-1 entry. The CCR5 inhibitor maraviroc has been shown in tissue culture to be able to repel cell-free virions from the cell surface into extracellular space. We hypothesized that cenicriviroc might exhibit a similar effect, and tested this using clinical samples from the Phase IIb study 652-2-202, by measuring rates of intracellular DNA decline. We also monitored viral RNA levels in culture fluids. METHODS: We infected PM-1 cells with CCR5-tropic HIV-1 BaL in the presence or absence of inhibitory concentrations of cenicriviroc (20 nM) or maraviroc (50 nM) or controls. Viral load levels and p24 were measured by ELISA, quantitative PCR and quantitative real-time reverse transcription PCR at 4 h post-infection. Frozen PBMC DNA samples from 30 patients with virological success in the Phase IIb study were studied, as were early and late reverse transcript levels. Docking studies compared binding between cenicriviroc/CCR5 and maraviroc/CCR5. RESULTS: Unlike maraviroc, cenicriviroc did not cause an increase in the amount of virus present in culture fluids at 4 h compared with baseline. The use of cenicriviroc did, however, result in lower levels of intracellular viral DNA after 4 h. Structural modelling indicates that cenicriviroc binds more deeply than maraviroc to the hydrophobic pocket of CCR5, providing an explanation for the absence of viral rebound with cenicriviroc. CONCLUSIONS: In contrast to maraviroc, cenicriviroc does not repel virus back into extracellular space. Differences in results may be due to superior binding of cenicriviroc to CCR5 compared with maraviroc.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Viral/analysis , HIV Infections/virology , HIV-1/isolation & purification , Imidazoles/pharmacology , Viral Load , Anti-HIV Agents/therapeutic use , Cell Line , Clinical Trials, Phase II as Topic , Culture Media , Enzyme-Linked Immunosorbent Assay , Extracellular Space/virology , HIV Infections/drug therapy , Humans , Imidazoles/therapeutic use , Leukocytes, Mononuclear/virology , Real-Time Polymerase Chain Reaction , Sulfoxides , Virus Cultivation
7.
Curr HIV Res ; 13(2): 90-7, 2015.
Article in English | MEDLINE | ID: mdl-25439235

ABSTRACT

The human immunodeficiency virus type 1 (HIV) eradication will require elimination of HIV infected cells. No antiretroviral treatments (ART) or vaccine approaches have been able to reduce significantly the level of HIV infected cells in peripheral blood. This inefficacy is generally explained by the presence of a major reservoir of latent HIV infected cells in the central nervous system (CNS) that would be a sanctuary where Cytotoxic T Lymphocytes (CTL) have no access and would refresh peripheral blood with activated HIV infected cells. In this review, the presence of a major reservoir in the CNS appears to be inconsistent with recent clinical studies measuring HIV DNA. The major reservoirs are gut tissue, rectal tissue and the peripheral blood where HIV infected cells survive in an environment containing CTL. Extracellular Tat might protect HIV infected cells from CTL due to its capacity to cross CTL membranes and trigger apoptosis. Evidences of Tat secretion from HIV infected cells are shown with the detection of Tat antibodies in different clinical studies. Presence of neutralizing Tat antibodies in cohorts of patients who were exposed to HIV but who are now seronegative is described. The conclusion of this review is that a vaccine eliciting neutralizing antibodies against Tat might significantly reduce the level of HIV infected cells, what ART or other vaccine approaches have been unable to achieve now. It could be a first step towards HIV eradication.


Subject(s)
Antibodies, Neutralizing/immunology , Central Nervous System/virology , HIV Infections/virology , HIV-1/pathogenicity , Virus Activation/physiology , tat Gene Products, Human Immunodeficiency Virus/physiology , AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Extracellular Space/virology , HIV Infections/blood , HIV Infections/immunology , HIV Seronegativity/immunology , Humans , Intestines/virology , T-Lymphocytes, Cytotoxic/virology , tat Gene Products, Human Immunodeficiency Virus/immunology
8.
Immunol Res ; 57(1-3): 268-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24293008

ABSTRACT

The evolution of early multicellular eukaryotes 400-500 million years ago required a defensive strategy against microbial invasion. Pore-forming proteins containing the membrane-attack-complex-perforin (MACPF) domain were selected as the most efficient means to destroy bacteria or virally infected cells. The mechanism of pore formation by the MACPF domain is distinctive in that pore formation is purely physical and unspecific. The MACPF domain polymerizes, refolds, and inserts itself into bilayer membranes or bacterial outer cell walls. The displacement of surface lipid/carbohydrate molecules by the polymerizing MACPF domain creates clusters of large, water-filled holes that destabilize the barrier function and provide access for additional anti-bacterial or anti-viral effectors to sensitive sites that complete the destruction of the invader via enzymatic or chemical attack. The highly efficient mechanism of anti-microbial defense by a combined physical and chemical strategy using pore-forming MACPF-proteins has been retargeted during evolution of vertebrates and mammals for three purposes: (1) to kill extracellular bacteria C9/polyC9 evolved in conjunction with complement, (2) to kill virus infected and cancer cells perforin-1/polyperforin-1 CTL evolved targeted by NK and CTL, and (3) to kill intracellular bacteria transmembrane perforin-2/putative polyperforin-2 evolved targeted by phagocytic and nonphagocytic cells. Our laboratory has been involved in the discovery and description of each of the three pore-formers that will be reviewed here.


Subject(s)
Immunity/physiology , Pore Forming Cytotoxic Proteins/physiology , Animals , Bacteria/immunology , Complement Membrane Attack Complex , Extracellular Space/immunology , Extracellular Space/microbiology , Extracellular Space/virology , Humans , Intracellular Space/immunology , Intracellular Space/microbiology , Neoplasms/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism
9.
PLoS Comput Biol ; 9(11): e1003372, 2013.
Article in English | MEDLINE | ID: mdl-24278009

ABSTRACT

Influenza A viruses are respiratory pathogens that cause seasonal epidemics with up to 500,000 deaths each year. Yet there are currently only two classes of antivirals licensed for treatment and drug-resistant strains are on the rise. A major challenge for the discovery of new anti-influenza agents is the identification of drug targets that efficiently interfere with viral replication. To support this step, we developed a multiscale model of influenza A virus infection which comprises both the intracellular level where the virus synthesizes its proteins, replicates its genome, and assembles new virions and the extracellular level where it spreads to new host cells. This integrated modeling approach recapitulates a wide range of experimental data across both scales including the time course of all three viral RNA species inside an infected cell and the infection dynamics in a cell population. It also allowed us to systematically study how interfering with specific steps of the viral life cycle affects virus production. We find that inhibitors of viral transcription, replication, protein synthesis, nuclear export, and assembly/release are most effective in decreasing virus titers whereas targeting virus entry primarily delays infection. In addition, our results suggest that for some antivirals therapy success strongly depends on the lifespan of infected cells and, thus, on the dynamics of virus-induced apoptosis or the host's immune response. Hence, the proposed model provides a systems-level understanding of influenza A virus infection and therapy as well as an ideal platform to include further levels of complexity toward a comprehensive description of infectious diseases.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , Influenza A virus/drug effects , Influenza, Human/virology , Models, Biological , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Cell Death , Computational Biology , Dogs , Extracellular Space/virology , Humans , Influenza A virus/physiology , Intracellular Space/virology , Madin Darby Canine Kidney Cells , Virus Internalization/drug effects
10.
PLoS One ; 8(7): e69450, 2013.
Article in English | MEDLINE | ID: mdl-23922713

ABSTRACT

During HIV pathogenesis, infected macrophages behave as "viral reservoirs" that accumulate and retain virions within dedicated internal Virus-Containing Compartments (VCCs). The nature of VCCs remains ill characterized and controversial. Using wild-type HIV-1 and a replication-competent HIV-1 carrying GFP internal to the Gag precursor, we analyzed the biogenesis and evolution of VCCs in primary human macrophages. VCCs appear roughly 14 hours after viral protein synthesis is detected, initially contain few motile viral particles, and then mature to fill up with virions that become packed and immobile. The amount of intracellular Gag, the proportion of dense VCCs, and the density of viral particles in their lumen increased with time post-infection. In contrast, the secretion of virions, their infectivity and their transmission to T cells decreased overtime, suggesting that HIV-infected macrophages tend to pack and retain newly formed virions into dense compartments. A minor proportion of VCCs remains connected to the plasma membrane overtime. Surprisingly, live cell imaging combined with correlative light and electron microscopy revealed that such connections can be transient, highlighting their dynamic nature. Together, our results shed light on the late phases of the HIV-1 cycle and reveal some of its macrophage specific features.


Subject(s)
Cell Compartmentation , Cell Membrane/virology , HIV-1/physiology , Macrophages/pathology , Macrophages/virology , Virion/physiology , Adult , Cell Membrane/ultrastructure , Extracellular Space/virology , HIV-1/ultrastructure , Humans , Macrophages/ultrastructure , Models, Biological , Time Factors , Virion/ultrastructure
11.
J Virol ; 87(17): 9441-51, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23804639

ABSTRACT

Histo-blood group antigens (HBGAs) have been suggested to be receptors or coreceptors for human noroviruses (HuNoVs) expressed on the intestinal epithelium. We isolated an enteric bacterium strain (SENG-6), closely related to Enterobacter cloacae, bearing HBGA-like substances from a fecal sample of a healthy individual by using a biopanning technique with anti-HBGA antibodies. The binding capacities of four genotypes of norovirus-like particles (NoVLPs) to Enterobacter sp. SENG-6 cells were confirmed by enzyme-linked immunosorbent assay (ELISA). Transmission electron microscopy demonstrated that NoVLPs bound mainly to extracellular polymeric substances (EPS) of Enterobacter sp. SENG-6, where the HBGA-like substances were localized. EPS that contained HBGA-like substances extracted from Enterobacter sp. SENG-6 was shown by enzyme-linked immunosorbent assay (ELISA) to be capable of binding to NoVLPs of a GI.1 wild-type strain (8fIIa) and a GII.6 strain that can recognize A antigen but not to an NoVLP GI.1 mutant strain (W375A) that loses the ability to bind to A antigen. Enzymatic cleavage of terminal N-acetyl-galactosamine residues in the bacterial EPS weakened bacterial EPS binding to the GI.1 wild-type strain (8fIIa). These results indicate that A-like substances in the bacterial EPS play a key role in binding to NoVLPs. Since the specific binding of HuNoVs to HBGA-positive enteric bacteria is likely to affect the transmission and infection processes of HuNoVs in their hosts and in the environment, further studies of human enteric bacteria and their binding capacity to HuNoVs will provide a new scientific platform for understanding interactions between two types of microbes that were previously regarded as biologically unrelated.


Subject(s)
Blood Group Antigens/physiology , Enterobacteriaceae/immunology , Enterobacteriaceae/virology , Norovirus/pathogenicity , Adsorption , Antigens, Bacterial/genetics , Antigens, Bacterial/physiology , Enterobacter/genetics , Enterobacter/immunology , Enterobacter/virology , Enterobacteriaceae/isolation & purification , Extracellular Space/immunology , Extracellular Space/virology , Feces/microbiology , Feces/virology , Humans , Molecular Sequence Data , Norovirus/immunology , Norovirus/physiology , Phylogeny , RNA, Bacterial/genetics , Virion/physiology , Virion/ultrastructure
12.
Biomaterials ; 34(13): 3511-22, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23415044

ABSTRACT

Recently, peptide drugs have evolved into mainstream therapeutics, representing a significant portion of the pharmaceutical market. However, their bioavailability remains to be improved compared with that of chemical drugs. Here, we screened and identified a new peptide, Ctry2459, from a scorpion venom peptide library that was proven to inhibit hepatitis C virus (HCV) infection via inactivating infectious viral particles. However, Ctry2459 cannot suppress established infection of HCV because of the poor cellular uptake and restriction of endosomes. Based on the molecular template of the Ctry2459 peptide, we designed two histidine-rich peptides (Ctry2459-H2 and Ctry2459-H3) with significantly enhanced cellular uptake and improved intracellular distribution. Moreover, the two mutated peptides, as well as the wild-type peptide Ctry2459, exhibited virucidal activities against HCV. In distinct contrast to the Ctry2459 peptide, the mutated peptides significantly suppressed the established HCV infection at the cellular level but demonstrated lower cytotoxic and hemolytic activities. Our work presents an effective design strategy for optimizing natural antiviral peptides and opens a new avenue for enhancing the bioavailability of peptide drugs.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Hepacivirus/drug effects , Peptides/pharmacology , Peptides/pharmacokinetics , Proteins/pharmacology , Acids/metabolism , Amino Acid Sequence , Antiviral Agents/analysis , Antiviral Agents/chemistry , Biological Availability , Cell Line, Tumor , Endosomes/drug effects , Endosomes/metabolism , Extracellular Space/drug effects , Extracellular Space/virology , Gene Library , Hemolysis/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/virology , Lysosomes/drug effects , Lysosomes/metabolism , Models, Biological , Molecular Sequence Data , Peptides/analysis , Peptides/chemistry , Protein Transport/drug effects , Proteins/chemistry , Proton-Translocating ATPases/metabolism , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacokinetics , Scorpion Venoms/pharmacology , Time Factors , Virus Inactivation/drug effects
13.
PLoS One ; 8(12): e83842, 2013.
Article in English | MEDLINE | ID: mdl-24386290

ABSTRACT

Gammaherpesvirinae, such as the human Epstein-Barr virus (EBV) and the Kaposi's sarcoma associated herpesvirus (KSHV) are highly prevalent pathogens that have been associated with several neoplastic diseases. As EBV and KSHV are host-range specific and replicate poorly in vitro, animal counterparts such as Murid herpesvirus-4 (MuHV-4) have been widely used as models. In this study, we used MuHV-4 in order to improve the knowledge about proteins that compose gammaherpesviruses virions. To this end, MuHV-4 extracellular virions were isolated and structural proteins were identified using liquid chromatography tandem mass spectrometry-based proteomic approaches. These analyses allowed the identification of 31 structural proteins encoded by the MuHV-4 genome which were classified as capsid (8), envelope (9), tegument (13) and unclassified (1) structural proteins. In addition, we estimated the relative abundance of the identified proteins in MuHV-4 virions by using exponentially modified protein abundance index analyses. In parallel, several host proteins were found in purified MuHV-4 virions including Annexin A2. Although Annexin A2 has previously been detected in different virions from various families, its role in the virion remains controversial. Interestingly, despite its relatively high abundance in virions, Annexin A2 was not essential for the growth of MuHV-4 in vitro. Altogether, these results extend previous work aimed at determining the composition of gammaherpesvirus virions and provide novel insights for understanding MuHV-4 biology.


Subject(s)
Extracellular Space/virology , Proteomics , Rhadinovirus/metabolism , Virion/metabolism , Animals , Capsid/metabolism , Cell Line , Cricetinae , Glycosylation , Mass Spectrometry , Viral Proteins/metabolism
14.
Cell Host Microbe ; 12(1): 109-16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22817992

ABSTRACT

Neutrophils contribute to pathogen clearance by producing neutrophil extracellular traps (NETs), which are genomic DNA-based net-like structures that capture bacteria and fungi. Although NETs also express antiviral factors, such as myeloperoxidase and α-defensin, the involvement of NETs in antiviral responses remains unclear. We show that NETs capture human immunodeficiency virus (HIV)-1 and promote HIV-1 elimination through myeloperoxidase and α-defensin. Neutrophils detect HIV-1 by Toll-like receptors (TLRs) TLR7 and TLR8, which recognize viral nucleic acids. Engagement of TLR7 and TLR8 induces the generation of reactive oxygen species that trigger NET formation, leading to NET-dependent HIV-1 elimination. However, HIV-1 counteracts this response by inducing C-type lectin CD209-dependent production of interleukin (IL)-10 by dendritic cells to inhibit NET formation. IL-10 suppresses the reactive oxygen species-dependent generation of NETs induced upon TLR7 and TLR8 engagement, resulting in disrupted NET-dependent HIV-1 elimination. Therefore, NET formation is an antiviral response that is counteracted by HIV-1.


Subject(s)
Extracellular Space/virology , HIV-1/pathogenicity , Host-Pathogen Interactions , Neutrophils/metabolism , Neutrophils/virology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dendritic Cells/virology , Extracellular Space/metabolism , Humans , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Neutrophils/cytology , Peroxidase/metabolism , Receptors, Cell Surface/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , alpha-Defensins/metabolism
15.
PLoS One ; 6(10): e26463, 2011.
Article in English | MEDLINE | ID: mdl-22028885

ABSTRACT

During viral infections, single- and double-stranded RNA (ssRNA and dsRNA) are recognized by the host and induce innate immune responses. The cellular enzyme ADAR-1 (adenosine deaminase acting on RNA-1) activation in virally infected cells leads to presence of inosine-containing RNA (Ino-RNA). Here we report that ss-Ino-RNA is a novel viral recognition element. We synthesized unmodified ssRNA and ssRNA that had 6% to16% inosine residues. The results showed that in primary human cells, or in mice, 10% ss-Ino-RNA rapidly and potently induced a significant increase in inflammatory cytokines, such as interferon (IFN)-ß (35 fold), tumor necrosis factor (TNF)-α (9.7 fold), and interleukin (IL)-6 (11.3 fold) (p<0.01). Flow cytometry data revealed a corresponding 4-fold increase in influx of neutrophils into the lungs by ss-Ino-RNA treatment. In our in vitro experiments, treatment of epithelial cells with ss-Ino-RNA reduced replication of respiratory syncytial virus (RSV). Interestingly, RNA structural analysis showed that ss-Ino-RNA had increased formation of secondary structures. Our data further revealed that extracellular ss-Ino-RNA was taken up by scavenger receptor class-A (SR-A) which activated downstream MAP Kinase pathways through Toll-like receptor 3 (TLR3) and dsRNA-activated protein kinase (PKR). Our data suggests that ss-Ino-RNA is an as yet undescribed virus-associated innate immune stimulus.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Immunity, Innate/drug effects , Inosine , RNA/chemistry , RNA/pharmacology , Respiratory Syncytial Viruses/drug effects , Animals , Antiviral Agents/metabolism , Base Sequence , Cell Line , Chemokines/biosynthesis , Chemokines/metabolism , Endocytosis , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Extracellular Space/drug effects , Extracellular Space/immunology , Extracellular Space/metabolism , Extracellular Space/virology , Humans , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Nucleic Acid Conformation , Protein Kinases/metabolism , RNA/metabolism , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/physiology , Scavenger Receptors, Class A/metabolism , Toll-Like Receptor 3/metabolism , Transcriptome/drug effects , Transcriptome/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Virus Replication/drug effects , Virus Replication/immunology
16.
PLoS One ; 6(10): e25115, 2011.
Article in English | MEDLINE | ID: mdl-21998635

ABSTRACT

Human cytomegalovirus (HCMV) UL77 gene encodes the essential protein UL77, its function is characterized in the present study. Immunoprecipitation identified monomeric and oligomeric pUL77 in HCMV infected cells. Immunostaining of purified virions and subviral fractions showed that pUL77 is a structural protein associated with capsids. In silico analysis revealed the presence of a coiled-coil motif (CCM) at the N-terminus of pUL77. Chemical cross-linking of either wild-type pUL77 or CCM deletion mutant (pUL77ΔCCM) implicated that CCM is critical for oligomerization of pUL77. Furthermore, co-immunoprecipitations of infected and transfected cells demonstrated that pUL77 interacts with the capsid-associated DNA packaging motor components, pUL56 and pUL104, as well as the major capsid protein. The ability of pUL77 to bind dsDNA was shown by an in vitro assay. Binding to certain DNA was further confirmed by an assay using biotinylated 36-, 250-, 500-, 1000-meric dsDNA and 966-meric HCMV-specific dsDNA designed for this study. The binding efficiency (BE) was determined by image processing program defining values above 1.0 as positive. While the BE of the pUL56 binding to the 36-mer bio-pac1 containing a packaging signal was 10.0 ± 0.63, the one for pUL77 was only 0.2±0.03. In contrast to this observation the BE of pUL77 binding to bio-500 bp or bio-1000 bp was 2.2 ± 0.41 and 4.9 ± 0.71, respectively. By using pUL77ΔCCM it was demonstrated that this protein could not bind to dsDNA. These data indicated that pUL77 (i) could form homodimers, (ii) CCM of pUL77 is crucial for oligomerization and (iii) could bind to dsDNA in a sequence independent manner.


Subject(s)
Cytomegalovirus , DNA, Viral/metabolism , Protein Multimerization , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Base Pairing , Capsid/metabolism , Computational Biology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , DNA Packaging , DNA, Viral/chemistry , DNA, Viral/genetics , Extracellular Space/virology , HEK293 Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Sequence Deletion , Viral Proteins/genetics , Virion/genetics , Virion/metabolism
17.
PLoS One ; 6(6): e20597, 2011.
Article in English | MEDLINE | ID: mdl-21687676

ABSTRACT

Protein-based subunit smallpox vaccines have shown their potential as effective alternatives to live virus vaccines in animal model challenge studies. We vaccinated mice with combinations of three different vaccinia virus (VACV) proteins (A33, B5, L1) and examined how the combined antibody responses to these proteins cooperate to effectively neutralize the extracellular virus (EV) infectious form of VACV. Antibodies against these targets were generated in the presence or absence of CpG adjuvant so that Th1-biased antibody responses could be compared to Th2-biased responses to the proteins with aluminum hydroxide alone, specifically with interest in looking at the ability of anti-B5 and anti-A33 polyclonal antibodies (pAb) to utilize complement-mediated neutralization in vitro. We found that neutralization of EV by anti-A33 or anti-B5 pAb can be enhanced in the presence of complement if Th1-biased antibody (IgG2a) is generated. Mechanistic differences found for complement-mediated neutralization showed that anti-A33 antibodies likely result in virolysis, while anti-B5 antibodies with complement can neutralize by opsonization (coating). In vivo studies found that mice lacking the C3 protein of complement were less protected than wild-type mice after passive transfer of anti-B5 pAb or vaccination with B5. Passive transfer of anti-B5 pAb or monoclonal antibody into mice lacking Fc receptors (FcRs) found that FcRs were also important in mediating protection. These results demonstrate that both complement and FcRs are important effector mechanisms for antibody-mediated protection from VACV challenge in mice.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Complement System Proteins/metabolism , Extracellular Space/virology , Receptors, Fc/metabolism , Vaccinia virus/immunology , Adjuvants, Immunologic/metabolism , Animals , CpG Islands/immunology , Extracellular Space/immunology , Extracellular Space/metabolism , Female , Immunization, Passive , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Opsonin Proteins/metabolism , Vaccination , Viral Proteins/immunology
18.
Mol Plant Microbe Interact ; 23(3): 263-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20121448

ABSTRACT

Plant viruses hijack endogenous host transport machinery to aid their intracellular spread. Here, we study the localization of the p7B, the membrane-associated viral movement protein (MP) of the Melon necrotic spot virus (MNSV), and also the potential involvement of the secretory pathway on the p7B targeting and intra- and intercellular virus movements. p7B fused to fluorescent proteins was located throughout the endoplasmic reticulum (ER) at motile Golgi apparatus (GA) stacks that actively tracked the actin microfilaments, and at the plasmodesmata (PD). Hence, the secretory pathway inhibitor, Brefeldin A (BFA), and the overexpression of the GTPase-defective mutant of Sar1p, Sar1[H74L], fully retained the p7B within the ER, revealing that the protein is delivered to PD in a BFA-sensitive and COPII-dependent manner. Disruption of the actin cytoskeleton with latrunculin B led to the accumulation of p7B in the ER, which strongly suggests that p7B is also targeted to the cell periphery in an actin-dependent manner. Remarkably, the local spread of the viral infection was significantly restricted either with the presence of BFA or under the overexpression of Sar1[H74L], thus revealing the involvement of an active secretory pathway in the intracellular movement of MNSV. Overall, these findings support a novel route for the intracellular transport of a plant virus led by the GA.


Subject(s)
Carmovirus/metabolism , Plant Viral Movement Proteins/metabolism , Secretory Pathway , Viral Proteins/metabolism , Biological Transport/drug effects , Brefeldin A/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carmovirus/genetics , Carmovirus/physiology , Cucurbitaceae/virology , Endoplasmic Reticulum/metabolism , Extracellular Space/virology , Golgi Apparatus/metabolism , Host-Pathogen Interactions , Intracellular Space/virology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Mutation , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Leaves/virology , Plant Viral Movement Proteins/genetics , Plasmodesmata/metabolism , Protein Transport/drug effects , Thiazolidines/pharmacology , Nicotiana/genetics , Nicotiana/metabolism , Nicotiana/virology , Viral Proteins/genetics
19.
J Immunol ; 184(6): 3242-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20173031

ABSTRACT

Human aging is characterized by expanded and altered adaptive immune responses to human CMV (HCMV). It is unclear whether this expansion has its origins in age-related homeostatic disturbances or viral reactivation, whether anti-CMV immune surveillance may still be effective, and what are the consequences of this expanded immune response for health and longevity. We conducted an observational cross-sectional study in groups of HCMV-seropositive subjects aged >or=65 y of variable health status to compare the intensity of Ab responses against HCMV with those against EBV and with CD4(+) and CD8(+) T cell proinflammatory effector responses directed to HCMV-derived pp65 and immediate-early protein 1 synthetic peptides. Ab responses to HCMV, but not to EBV, and anti-HCMV CD4(+), but not CD8(+), T cell responses were more intense in elderly subjects aged >or=85 y in poor health and were inversely correlated with markers of functional activity and cognitive function. Therefore, humoral and CD4(+) T cell anti-HCMV responses were specifically intensified in advanced aging associated with comorbidity and cognitive and functional impairments. Such a distinctive pattern of adaptive immunity indicates that immune responses targeting the extracellular phase of HCMV are increased in these elderly subjects and could represent an indirect effect of localized and undetectable HCMV reactivation. This study demonstrates that the oldest subjects in poor health with physical and mental impairment express intense functional immune responses to extracellular HCMV and suggests that they may be at risk for direct pathogenic effects by HCMV reactivation as well as indirect pathogenic effects linked to proinflammatory anti-HCMV effector responses.


Subject(s)
Adaptive Immunity , Cognition Disorders/immunology , Cognition Disorders/psychology , Cytomegalovirus/immunology , Extracellular Space/immunology , Extracellular Space/virology , Aged , Aged, 80 and over , Antibodies, Viral/biosynthesis , Brief Psychiatric Rating Scale , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cognition Disorders/epidemiology , Comorbidity , Cross-Sectional Studies , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immediate-Early Proteins/chemical synthesis , Immediate-Early Proteins/immunology , Inflammation Mediators/physiology , Male , Phosphoproteins/chemical synthesis , Phosphoproteins/immunology , Viral Matrix Proteins/chemical synthesis , Viral Matrix Proteins/immunology , Virus Activation/immunology
20.
Cell Commun Adhes ; 15(5-6): 385-95, 2009.
Article in English | MEDLINE | ID: mdl-19306227

ABSTRACT

Analyses of images of the cell-to-substratum region of contact have been carried out by the means of total internal reflection fluorescence (TIRF) microscopy during both the formation and the dissolution of cellular contacts. The evolutions of the cellular contacts are visualized during the adhesion process under normal and virus-infected intracellular conditions, and during the lift-off process under various toxicities of the extracellular medium fluid. Then, propositions are developed for quantifying the cell viabilities by estimating the increase in the area of contact for the adhesion process and by specifying the maximum intensity of the TIRF image for the lift-off process.


Subject(s)
Microscopy, Fluorescence/methods , Adenoviridae/physiology , Cell Adhesion , Extracellular Space/virology , HEK293 Cells , Humans , Intracellular Space/virology , Microscopy, Fluorescence/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...