Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
J Virol ; 96(14): e0066522, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35762760

ABSTRACT

Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients, and its recent propagation in cell culture has opened the possibility to study its biology. Unlike classical human astroviruses, VA1 growth was found to be independent of trypsin during virus replication in vitro. In this work, we show that despite its independence on trypsin activation for cell infection, the VA1 capsid precursor protein, of 86 kDa (VP86), is processed intracellularly, and this proteolytic processing is important for astrovirus VA1 infectivity. Antibodies raised against different regions of the capsid precursor showed that the polyprotein can be processed starting at either its amino- or carboxy-terminal end, and they allowed us to identify those proteins of about 33 (VP33) and 38 (VP38) kDa constitute the core and the spike proteins of the mature infectious virus particles, respectively. The amino-terminal end of the spike protein was found to be Thr-348. Whether the protease involved in intracellular cleavage of the capsid precursor is of viral or cellular origin remains to be determined, but the cleavage is independent of caspases. Also, trypsin is able to degrade the capsid precursor but has no effect on VP33 and VP38 proteins when assembled into virus particles. These studies provide the basis for advancement of the knowledge of astrovirus VA1 cell entry and replication. IMPORTANCE Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients. Its recent propagation in cell culture has facilitated the study of its biology. In this work, we show that despite the ability of this virus to grow in the absence of trypsin, a marked feature of human classical astroviruses, the capsid precursor protein of astrovirus VA1 is cleaved intracellularly to yield the mature infectious particles, formed by two polypeptides, VP33 that constitutes the core domain of the virus particle, and VP38 that forms the spike of the virus. These studies provide a platform to advance our knowledge on astrovirus VA1 cell entry and replication.


Subject(s)
Astroviridae Infections , Capsid Proteins , Mamastrovirus , Protein Precursors , Astroviridae Infections/virology , Caco-2 Cells , Capsid/metabolism , Capsid Proteins/metabolism , Humans , Intracellular Space/virology , Mamastrovirus/physiology , Protein Precursors/metabolism , Trypsin/metabolism
2.
PLoS Pathog ; 17(1): e1009275, 2021 01.
Article in English | MEDLINE | ID: mdl-33513206

ABSTRACT

Filoviruses, such as the Ebola virus (EBOV) and Marburg virus (MARV), are causative agents of sporadic outbreaks of hemorrhagic fevers in humans. To infect cells, filoviruses are internalized via macropinocytosis and traffic through the endosomal pathway where host cathepsin-dependent cleavage of the viral glycoproteins occurs. Subsequently, the cleaved viral glycoprotein interacts with the late endosome/lysosome resident host protein, Niemann-Pick C1 (NPC1). This interaction is hypothesized to trigger viral and host membrane fusion, which results in the delivery of the viral genome into the cytoplasm and subsequent initiation of replication. Some studies suggest that EBOV viral particles activate signaling cascades and host-trafficking factors to promote their localization with host factors that are essential for entry. However, the mechanism through which these activating signals are initiated remains unknown. By screening a kinase inhibitor library, we found that receptor tyrosine kinase inhibitors potently block EBOV and MARV GP-dependent viral entry. Inhibitors of epidermal growth factor receptor (EGFR), tyrosine protein kinase Met (c-Met), and the insulin receptor (InsR)/insulin like growth factor 1 receptor (IGF1R) blocked filoviral GP-mediated entry and prevented growth of replicative EBOV in Vero cells. Furthermore, inhibitors of c-Met and InsR/IGF1R also blocked viral entry in macrophages, the primary targets of EBOV infection. Interestingly, while the c-Met and InsR/IGF1R inhibitors interfered with EBOV trafficking to NPC1, virus delivery to the receptor was not impaired in the presence of the EGFR inhibitor. Instead, we observed that the NPC1 positive compartments were phenotypically altered and rendered incompetent to permit viral entry. Despite their different mechanisms of action, all three RTK inhibitors tested inhibited virus-induced Akt activation, providing a possible explanation for how EBOV may activate signaling pathways during entry. In sum, these studies strongly suggest that receptor tyrosine kinases initiate signaling cascades essential for efficient post-internalization entry steps.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Chlorocebus aethiops , Ebolavirus/genetics , Endocytosis , Endosomes/metabolism , Endosomes/virology , Host-Pathogen Interactions , Humans , Intracellular Space/virology , Lysosomes/metabolism , Protein Transport , Protein-Tyrosine Kinases/genetics , Vero Cells , Virion , Virus Internalization , Virus Replication
3.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204285

ABSTRACT

Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.


Subject(s)
Host-Pathogen Interactions , Organelles/metabolism , Animals , Biological Transport , Biomarkers , Energy Metabolism , Host-Parasite Interactions , Humans , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Intracellular Space/virology , Organelles/microbiology , Organelles/parasitology , Organelles/ultrastructure , Phagocytosis
4.
J Viral Hepat ; 27(3): 261-269, 2020 03.
Article in English | MEDLINE | ID: mdl-31670859

ABSTRACT

The decay rate of hepatitis C virus (HCV)-infected cells during therapy has been used to determine the duration of treatment needed to attain a sustained virologic response, but with direct-acting anti-virals (DAA), this rate has been difficult to estimate. Here, we show that it is possible to estimate it, by simultaneously analysing the viral load and alanine aminotransferase (ALT) kinetics during combination DAA therapy. We modelled the HCV RNA and ALT serum kinetics in 26 patients with chronic HCV genotype 1b infection, under four different sofosbuvir-based combination treatments. In all patients, ALT decayed exponentially to a set point in the normal range by 1-3 weeks after initiation of therapy. The model indicates that the ALT decay rate during the first few weeks after initiation of therapy reflects the death rate of infected cells, with an estimated median half-life of 2.5 days in this patient population. This information allows independent estimation of the rate of loss of intracellular replication complexes during therapy. Our model also predicts that the final ALT set point is not related to the release of ALT by dying HCV-infected cells. Using ALT data, one can separately obtain information about the rate of 'cure' of HCV-infected cells versus their rate of death, something not possible when analysing only HCV RNA data. This information can be used to compare the effects of different DAA combinations and to rationally evaluate their anti-viral effects.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Intracellular Space/virology , Models, Theoretical , RNA, Viral/genetics , Virus Replication , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Genotype , Hepacivirus/physiology , Humans , Sustained Virologic Response , Viral Load
5.
J Cell Sci ; 130(1): 260-268, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27026525

ABSTRACT

Positive-strand RNA viruses, which can be devastating pathogens in humans, animals and plants, replicate their genomes on intracellular membranes. Here, we describe the three-dimensional ultrastructural organization of a tombusvirus replicase in yeast, a valuable model for exploring virus-host interactions. We visualized the intracellular distribution of a viral replicase protein using metal-tagging transmission electron microscopy, a highly sensitive nanotechnology whose full potential remains to be developed. These three-dimensional images show how viral replicase molecules are organized when they are incorporated into the active domains of the intracellular replication compartment. Our approach provides a means to study protein activation mechanisms in cells and to identify targets for new antiviral compounds.


Subject(s)
Imaging, Three-Dimensional , Intracellular Space/virology , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Tombusvirus/physiology , Virus Assembly , Antibodies/metabolism , Metallothionein/metabolism , Models, Biological , RNA, Double-Stranded/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae/virology , Tombusvirus/ultrastructure , Tomography , Virus Replication
6.
J Theor Biol ; 473: 20-27, 2019 07 21.
Article in English | MEDLINE | ID: mdl-31004612

ABSTRACT

Even in the presence of a successful combination therapy stalling the progress of AIDS, developing a cure for this disease is still an open question. One of the major steps towards a cure would be to be able to eradicate latent HIV reservoirs present in patients. During the last decade, multiple findings point to the dominant role of the viral protein Tat in the establishment of latency. Here we present a mathematical study to understand the potential role of Tat inhibitors as virus-suppressing agents. For this aim, we implemented a computational model that reproduces intracellular dynamics. Simulating an HIV-infected cell and its intracellular feedback we observed that removing Tat protein from the system via inhibitors resulted in a temporary and reversible viral suppression. In contrast, we observed that compounds that interact with Tat protein and disrupt the integrated viral genome produced a more permanent viral suppression.


Subject(s)
Antiviral Agents/pharmacology , HIV/physiology , Models, Biological , Virus Latency/drug effects , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Computer Simulation , Gene Expression Regulation, Viral/drug effects , HIV/drug effects , HIV/genetics , Intracellular Space/virology , Time Factors , tat Gene Products, Human Immunodeficiency Virus/metabolism
7.
Proc Natl Acad Sci U S A ; 112(29): 8887-92, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26195777

ABSTRACT

In the present article we examine clonality in virus evolution. Most viruses retain an active recombination machinery as a potential means to initiate new levels of genetic exploration that go beyond those attainable solely by point mutations. However, despite abundant recombination that may be linked to molecular events essential for genome replication, herein we provide evidence that generation of recombinants with altered biological properties is not essential for the completion of the replication cycles of viruses, and that viral lineages (near-clades) can be defined. We distinguish mechanistically active but inconsequential recombination from evolutionarily relevant recombination, illustrated by episodes in the field and during experimental evolution. In the field, recombination has been at the origin of new viral pathogens, and has conferred fitness advantages to some viruses once the parental viruses have attained a sufficient degree of diversification by point mutations. In the laboratory, recombination mediated a salient genome segmentation of foot-and-mouth disease virus, an important animal pathogen whose genome in nature has always been characterized as unsegmented. We propose a model of continuous mutation and recombination, with punctuated, biologically relevant recombination events for the survival of viruses, both as disease agents and as promoters of cellular evolution. Thus, clonality is the standard evolutionary mode for viruses because recombination is largely inconsequential, since the decisive events for virus replication and survival are not dependent on the exchange of genetic material and formation of recombinant (mosaic) genomes.


Subject(s)
Biological Evolution , Intracellular Space/virology , Polyploidy , Viruses/genetics , Animals , Clone Cells , Foot-and-Mouth Disease Virus/genetics , Models, Biological , Recombination, Genetic/genetics , Terminology as Topic
8.
J Theor Biol ; 435: 229-237, 2017 12 21.
Article in English | MEDLINE | ID: mdl-28943403

ABSTRACT

Bacteriophage proteins are viruses that can significantly impact on the functioning of bacteria and can be used in phage based therapy. The functioning of Bacteriophage in the host bacteria depends on its location in those host cells. It is very important to know the subcellular location of the phage proteins in a host cell in order to understand their working mechanism. In this paper, we propose iPHLoc-ES, a prediction method for subcellular localization of bacteriophage proteins. We aim to solve two problems: discriminating between host located and non-host located phage proteins and discriminating between the locations of host located protein in a host cell (membrane or cytoplasm). To do this, we extract sets of evolutionary and structural features of phage protein and employ Support Vector Machine (SVM) as our classifier. We also use recursive feature elimination (RFE) to reduce the number of features for effective prediction. On standard dataset using standard evaluation criteria, our method significantly outperforms the state-of-the-art predictor. iPHLoc-ES is readily available to use as a standalone tool from: https://github.com/swakkhar/iPHLoc-ES/ and as a web application from: http://brl.uiu.ac.bd/iPHLoc-ES/.


Subject(s)
Bacteriophages/chemistry , Cell Compartmentation , Support Vector Machine/standards , Viral Proteins/metabolism , Evolution, Molecular , Host-Pathogen Interactions , Intracellular Space/virology , Models, Biological , Viral Proteins/genetics
9.
Immunol Rev ; 256(1): 300-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117829

ABSTRACT

The human immunodeficiency virus-1 (HIV-1) infects helper CD4(+) T cells, and causes CD4(+) T-cell depletion and immunodeficiency. In the past 30 years, significant progress has been made in antiretroviral therapy, and the disease has become manageable. Nevertheless, an effective vaccine is still nowhere in sight, and a cure or a functional cure awaits discovery. Among possible curative therapies, traditional antiretroviral therapy, mostly targeting viral proteins, has been proven ineffective. It is possible that targeting HIV-dependent host cofactors may offer alternatives, both for preventing HIV transmission and for forestalling disease progression. Recently, the actin cytoskeleton and its regulators in blood CD4(+) T cells have emerged as major host cofactors that could be targeted. The novel concept that the cortical actin is a barrier to viral entry and early post-entry migration has led to the nascent model of virus-host interaction at the cortical actin layer. Deciphering the cellular regulatory pathways has manifested exciting prospects for future therapeutics. In this review, we describe the study of HIV interactions with actin cytoskeleton. We also examine potential pharmacological targets that emerge from this interaction. In addition, we briefly discuss several actin pathway-based anti-HIV drugs that are currently in development or testing.


Subject(s)
Actins/metabolism , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/physiology , Receptors, Chemokine/metabolism , Signal Transduction , Actin Cytoskeleton/metabolism , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Biological Transport , Carrier Proteins/metabolism , Chemotaxis/immunology , GTP-Binding Proteins/metabolism , HIV Infections/drug therapy , HIV-1/drug effects , Humans , Intracellular Space/metabolism , Intracellular Space/virology , Protein Binding , Receptors, Chemokine/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
10.
Mol Cell Probes ; 30(4): 231-237, 2016 08.
Article in English | MEDLINE | ID: mdl-27328610

ABSTRACT

Human metapneumovirus (hMPV) is a prevalent pathogen worldwide and causes various respiratory infections. Although it is a critical pathogen in pediatric patients, it is unclear how it enters host cells. In this study, we focused on hMPV cell entry using two kinds of cell lines (Vero E6 and LLC-MK2), which are most commonly used for isolating and propagating for hMPV, and we used fluorescent dyes to label the virus particles and monitored how they enter the host cell in real time. We found that endocytosis was the predominant pathway by which hMPV entered host cells. When the virus particles were traced inside host cells, we found that a low intracellular pH was needed for intracellular fusion in LLC-MK2 cells.


Subject(s)
Endocytosis , Host-Pathogen Interactions , Metapneumovirus/physiology , Virus Internalization , Animals , Chlorocebus aethiops , Humans , Hydrogen-Ion Concentration , Intracellular Space/virology , LLC-PK1 Cells , Membrane Fusion , Swine , Vero Cells , Viral Proteins/metabolism
11.
J Theor Biol ; 366: 91-102, 2015 Feb 07.
Article in English | MEDLINE | ID: mdl-25476731

ABSTRACT

UNLABELLED: The progression of acute hepatitis B virus (HBV) to chronic infection or clearance is highly dependent on the host immune response composed of cytolytic (CTL) and non-cytolytic (non-CTL) effects. Cytolytic processes induce hepatocyte killing while non-CTL processes inhibit intracellular replication. Both effects are widely recognized and accepted. However, there are uncertainties about the assistance provided by either the loss of covalently circular closed DNA (cccDNA) during cell proliferation or the emergence of refractory cells to immune mediated clearance. We developed an agent-based mathematical model and tested the relative roles of different mechanisms of the immune system in the clearance of acute HBV infection. HBV viremia clearance time and hepatocyte turnover (HT) were used as the two major criteria in determining reasonable outcomes. Modelling results in 90% of cells containing between 1 and 17 cccDNA copies and normally distributed at the peak of infection. Variations in p36 levels, responsible for determining export of virions or recirculation to amplify cccDNA numbers, have a much greater impact on mean cccDNA level/cell at peak viremia than virus infectivity and cccDNA half-life. A strong CTL effect alone failed to clear infection with HT ≈ 10. Acute infection clearance was possible with combined CTL and non-CTL effects along with complete loss of intracellular viral components during cell proliferation resulting in the desired range of HT (0.7-1). The emergence of cells refractory to infection can reduce HT by up to 90%. However their impact was less effective than complete loss of intracellular viral components during cell proliferation. CONCLUSION: the existence of refractory cells is not necessary when there is complete loss of intracellular quantities during cell proliferation but is essential with only partial clearance.


Subject(s)
Computer Simulation , Hepatitis B virus/physiology , Hepatitis B/immunology , Hepatitis B/virology , Acute Disease , Adaptive Immunity , Animals , Antiviral Agents/therapeutic use , Cell Proliferation , Cytotoxicity, Immunologic , DNA, Circular/genetics , DNA, Viral/genetics , Hepatitis B/pathology , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Hepatocytes/pathology , Hepatocytes/virology , Humans , Intracellular Space/virology , Viremia/immunology , Viremia/virology , Virion/pathogenicity , Virus Replication
12.
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
13.
Proteomics ; 13(22): 3309-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24115376

ABSTRACT

Cross-species transmissions of swine influenza viruses (SIVs) raise great public health concerns. In this study, subcellular proteomic profiles of human A549 cells inoculated with H3N2 subtype SIV were used to characterize dynamic cellular responses to infection. By 2DE and MS, 27 differentially expressed (13 upregulated, 14 downregulated) cytoplasmic proteins and 20 differentially expressed (13 upregulated, 7 downregulated) nuclear proteins were identified. Gene ontology analysis suggested that these differentially expressed proteins were mainly involved in cell death, stress response, lipid metabolism, cell signaling, and RNA PTMs. Moreover, 25 corresponding genes of the differentially expressed proteins were quantitated by real time RT-PCR to examine the transcriptional profiles between mock- and virus-infected A549 cells. Western blot analysis confirmed that changes in abundance of identified cellular proteins heterogeneous nuclear ribonucleoprotein (hnRNP) U, hnRNP C, ALDH1A1, tryptophanyl-tRNA synthetase, IFI35, and HSPB1 in H3N2 SIV-infected cells were consistent with results of 2DE analysis. By confocal microscopy, nucleus-to-cytoplasm translocation of hnRNP C and colocalization between the viral nonstructural protein 1 and hnRNP C as well as N-myc (and STAT) interactor were observed upon infection. Ingenuity Pathway Analysis revealed that cellular proteins altered during infection were grouped mainly into NFκB and interferon signaling networks. Collectively, these identified subcellular constituents provide an important framework for understanding host/SIV interactions and underlying mechanisms of SIV cross-species infection and pathogenesis.


Subject(s)
Host-Pathogen Interactions/physiology , Influenza A Virus, H3N2 Subtype/physiology , Intracellular Space/chemistry , Intracellular Space/virology , Proteome/analysis , Amino Acid Sequence , Blotting, Western , Cell Line , Electrophoresis, Gel, Two-Dimensional , Humans , Intracellular Space/metabolism , Mass Spectrometry , Microscopy, Confocal , Molecular Sequence Data , Proteins/analysis , Proteins/chemistry , Proteins/classification , Proteins/metabolism , Proteome/chemistry , Proteome/metabolism , Transcriptome
14.
Environ Microbiol ; 15(8): 2306-18, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23489642

ABSTRACT

Microbes drive the biogeochemical cycles that fuel planet Earth, and their viruses (phages) alter microbial population structure, genome repertoire, and metabolic capacity. However, our ability to understand and quantify phage-host interactions is technique-limited. Here, we introduce phageFISH - a markedly improved geneFISH protocol that increases gene detection efficiency from 40% to > 92% and is optimized for detection and visualization of intra- and extracellular phage DNA. The application of phageFISH to characterize infection dynamics in a marine podovirus-gammaproteobacterial host model system corroborated classical metrics (qPCR, plaque assay, FVIC, DAPI) and outperformed most of them to reveal new biology. PhageFISH detected both replicating and encapsidated (intracellular and extracellular) phage DNA, while simultaneously identifying and quantifying host cells during all stages of infection. Additionally, phageFISH allowed per-cell relative measurements of phage DNA, enabling single-cell documentation of infection status (e.g. early vs late stage infections). Further, it discriminated between two waves of infection, which no other measurement could due to population-averaged signals. Together, these findings richly characterize the infection dynamics of a novel model phage-host system, and debut phageFISH as a much-needed tool for studying phage-host interactions in the laboratory, with great promise for environmental surveys and lineage-specific population ecology of free phages.


Subject(s)
Bacteriophages/genetics , Host-Pathogen Interactions , Intracellular Space/virology , Podoviridae/physiology , Pseudoalteromonas/virology , Virology/methods , Reproducibility of Results , Seawater/microbiology , Seawater/virology
15.
Plant Biotechnol J ; 10(4): 410-21, 2012 May.
Article in English | MEDLINE | ID: mdl-22260326

ABSTRACT

Human papillomavirus 8 (HPV-8), one of the high-risk cutaneous papillomaviruses (cHPVs), is associated with epidermodysplasia verruciformis and nonmelanoma skin cancer in immuno-compromised individuals. Currently, no vaccines against cHPVs have been reported; however, recent studies on cross-neutralizing properties of their capsid proteins (CP) have fostered an interest in vaccine production against these viruses. We examined the potential of producing HPV-8 major CP L1 in Nicotiana benthamiana by agroinfiltration of different transient expression vectors: (i) the binary vector pBIN19 with or without silencing suppressor constructs, (ii) the nonreplicating Cowpea mosaic virus-derived expression vector pEAQ-HT and (iii) a replicating Tobacco mosaic virus (TMV)-based vector alone or with signal peptides. Although HPV-8 L1 was successfully expressed using pEAQ-HT and TMV, a 15-fold increase was obtained with pEAQ-HT. In contrast, no L1 protein could be immune detected using pBIN19 irrespective of whether silencing suppressors were coexpressed, although such constructs were required for identifying L1-specific transcripts. A fourfold yield increase in L1 expression was obtained when 22 C-terminal amino acids were deleted (L1ΔC22), possibly eliminating a nuclear localization signal. Electron microscopy showed that plant-made HPV-8 L1 proteins assembled in appropriate virus-like particles (VLPs) of T = 1 or T = 7 symmetry. Ultrathin sections of L1ΔC22-expressing cells revealed their accumulation in the cytoplasm in the form of VLPs or paracrystalline arrays. These results show for the first time the production and localization of HPV-8 L1 protein in planta and its assembly into VLPs representing promising candidate for potential vaccine production.


Subject(s)
Capsid Proteins/biosynthesis , Capsid Proteins/isolation & purification , Gene Expression , Genetic Techniques , Nicotiana/metabolism , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Blotting, Northern , Capsid Proteins/ultrastructure , DNA, Bacterial/genetics , Genetic Vectors/genetics , Humans , Immunoblotting , Intracellular Space/metabolism , Intracellular Space/virology , Mutant Proteins/metabolism , Oncogene Proteins, Viral/ultrastructure , Plant Exudates/metabolism , Recombinant Proteins/ultrastructure , Subcellular Fractions/virology , Virion/metabolism , Virion/ultrastructure
16.
J Immunol ; 185(2): 1093-102, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20530255

ABSTRACT

Human CMV (HCMV) encodes multiple genes that control NK cell activation and cytotoxicity. Some of these HCMV-encoded gene products modulate NK cell activity as ligands expressed at the cell surface that engage inhibitory NK cell receptors, whereas others prevent the infected cell from upregulating ligands that bind to activating NK cell receptors. A major activating NKR is the homodimeric NKG2D receptor, which has eight distinct natural ligands in humans. It was shown that HCMV is able to prevent the surface expression of five of these ligands (MIC A/B and ULBP1, 2, and 6). In this article, we show that the HCMV gene product UL142 can prevent cell surface expression of ULBP3 during infection. We further show that UL142 interacts with ULBP3 and mediates its intracellular retention in a compartment that colocalizes with markers of the cis-Golgi complex. In doing so, UL142 prevents ULBP3 trafficking to the surface and protects transfected cells from NK-mediated cytotoxicity. This is the first description of a viral gene able to mediate downregulation of ULBP3.


Subject(s)
Cytomegalovirus/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/genetics , Viral Proteins/genetics , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Cytomegalovirus/genetics , Cytotoxicity, Immunologic/immunology , Fibroblasts/cytology , Fibroblasts/virology , GPI-Linked Proteins , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Space/metabolism , Intracellular Space/virology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Protein Transport , Recombinant Fusion Proteins/genetics , Transfection , Viral Proteins/metabolism
17.
J Gen Virol ; 92(Pt 8): 1832-1842, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21508188

ABSTRACT

The influenza A virus non-structural protein 1 (NS1) is a multifunctional protein and an important virulence factor. It is composed of two well-characterized domains linked by a short, but not well crystallographically defined, region of unknown function. To study the possible function of this region, we introduced alanine substitutions to replace the two highly conserved leucine residues at amino acid positions 69 and 77. The mutant L69,77A NS1 protein retained wild-type (WT)-comparable binding capabilities to dsRNA, cleavage and polyadenylation specificity factor 30 and the p85ß subunit of PI3K. A mutant influenza A virus expressing the L69,77A NS1 protein was generated using reverse genetics. L69,77A NS1 virus infection induced significantly higher levels of beta interferon (IFN-ß) expression in Madin-Darby canine kidney (MDCK) cells compared with WT NS1 virus. In addition, the replication rate of the L69,77A NS1 virus was substantially lower in MDCK cells but not in Vero cells compared with the WT virus, suggesting that the L69,77A NS1 protein does not fully antagonize IFN during viral replication. L69,77A NS1 virus infection was not able to activate the PI3K/Akt anti-apoptotic pathway, suggesting that the mutant NS1 protein may not be localized such that it has access to p85ß in vivo during infection, which was supported by the altered subcellular localization pattern of the mutant NS1 compared with WT NS1 after transfection or virus infection. Our data demonstrate that this linker region between the two domains is critical for the functions of the NS1 protein during influenza A virus infection, possibly by determining the protein's correct subcellular localization.


Subject(s)
Amino Acid Substitution , Influenza A virus/physiology , Influenza, Human/virology , Intracellular Space/virology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Amino Acid Motifs , Animals , Cell Line , Dogs , Humans , Influenza A virus/chemistry , Influenza A virus/genetics , Influenza, Human/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Protein Transport , Viral Nonstructural Proteins/genetics
18.
Immunol Cell Biol ; 89(2): 173-82, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20603636

ABSTRACT

MxA is an antiviral protein induced by interferon (IFN)-α/ß that is known to inhibit the replication of many RNA viruses. In these experiments, the 76-kDa MxA protein expressed in IFN-α-treated cells was shown to have antiviral activity against herpes simplex virus-1 (HSV-1), a human DNA virus. However, MxA was expressed as a 56-kDa protein in HSV-1-infected cells in the absence of IFN-α. This previously unrecognized MxA isoform was produced from an alternatively spliced MxA transcript that had a deletion of Exons 14-16 and a frame shift altering the C-terminus. The variant MxA (varMxA) isoform was associated with HSV-1 regulatory proteins and virions in nuclear replication compartments. varMxA expression enhanced HSV-1 infection as shown by a reduction in infectious virus titers from cells in which MxA had been inhibited by RNA interference and by an increase in HSV-1 titers when the 56-kDa varMxA was expressed constitutively. Thus, the human MxA gene encodes two MxA isoforms, which are expressed differentially depending on whether the stimulus is IFN-α or HSV-1. These findings show that alternative splicing of cellular mRNA can result in expression of a novel isoform of a host defense gene that supports instead of restricting viral infection.


Subject(s)
GTP-Binding Proteins/genetics , Herpesvirus 1, Human/physiology , Virus Replication/physiology , Alternative Splicing/drug effects , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/virology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fibroblasts/virology , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/ultrastructure , Humans , Interferon-alpha/pharmacology , Intracellular Space/drug effects , Intracellular Space/virology , Molecular Sequence Data , Myxovirus Resistance Proteins , Protein Biosynthesis/drug effects , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Virion/drug effects , Virion/physiology , Virus Replication/drug effects
19.
J Virol ; 84(5): 2294-303, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20032189

ABSTRACT

The filovirus VP40 protein is capable of budding from mammalian cells in the form of virus-like particles (VLPs) that are morphologically indistinguishable from infectious virions. Ebola virus VP40 (eVP40) contains well-characterized overlapping L domains, which play a key role in mediating efficient virus egress. L domains represent only one component required for efficient budding and, therefore, there is a need to identify and characterize additional domains important for VP40 function. We demonstrate here that the (96)LPLGVA(101) sequence of eVP40 and the corresponding (84)LPLGIM(89) sequence of Marburg virus VP40 (mVP40) are critical for efficient release of VP40 VLPs. Indeed, deletion of these motifs essentially abolished the ability of eVP40 and mVP40 to bud as VLPs. To address the mechanism by which the (96)LPLGVA(101) motif of eVP40 contributes to egress, a series of point mutations were introduced into this motif. These mutants were then compared to the eVP40 wild type in a VLP budding assay to assess budding competency. Confocal microscopy and gel filtration analyses were performed to assess their pattern of intracellular localization and ability to oligomerize, respectively. Our results show that mutations disrupting the (96)LPLGVA(101) motif resulted in both altered patterns of intracellular localization and self-assembly compared to wild-type controls. Interestingly, coexpression of either Ebola virus GP-WT or mVP40-WT with eVP40-DeltaLPLGVA failed to rescue the budding defective eVP40-DeltaLPLGVA mutant into VLPs; however, coexpression of eVP40-WT with mVP40-DeltaLPLGIM successfully rescued budding of mVP40-DeltaLPLGIM into VLPs at mVP40-WT levels. In sum, our findings implicate the LPLGVA and LPLGIM motifs of eVP40 and mVP40, respectively, as being important for VP40 structure/stability and budding.


Subject(s)
Amino Acid Motifs/genetics , Ebolavirus/genetics , Marburgvirus/genetics , Nucleoproteins/genetics , Viral Core Proteins/genetics , Virion , Animals , Cell Line , Humans , Intracellular Space/virology , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Alignment , Sequence Homology, Amino Acid , Virion/genetics , Virion/metabolism , Virion/ultrastructure , Virus Assembly/genetics , Virus Release
20.
J Virol ; 84(22): 11858-65, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20844052

ABSTRACT

Highly pathogenic avian influenza viruses of subtype H7N1 that emerged during an outbreak in 1999 and 2000 in Italy differ from their low-pathogenicity precursor viruses by changes in several genes, including three mutations in the NS1 protein. Two of them involve amino acid exchanges located within or closely adjacent to the nuclear export signal of NS1. The third mutation resulted in a new stop codon and thereby a C-terminal truncation of the NS1 protein of the highly pathogenic viruses. To find out whether these mutations contribute to the phenotypic differences between the highly pathogenic and low pathogenic viruses, we generated recombinants of the highly pathogenic A/ostrich/Italy/984/00 strain that contained the nuclear export signal and/or the extended C terminus of NS1 of a low pathogenic virus (A/chicken/Italy/1082/99). Using these recombinants we could demonstrate that replication rate and spread of infection in chicken fibroblast cultures, as well as infectivity for chicken embryos is reduced, whereas the mean death time for chicken embryos is increased, when the highly pathogenic virus acquires the NS1 motifs of the low pathogenic virus. Analysis of beta interferon transcription in chicken fibroblasts infected with the recombinants revealed that the mutations observed in the nuclear export signal of the highly pathogenic viruses were responsible for the enhanced interferon antagonism of these viruses. Cell fractionation and immunofluorescence studies in chicken fibroblasts showed that the nuclear export signal of the highly pathogenic viruses is responsible for cytoplasmic accumulation of NS1, whereas the C-terminal truncation promotes transport into the nucleoli. Comparative analysis in human A549 cells indicated that intracellular distribution of NS1 is host specific. Taken together, these observations support the concept that compartmentalization of NS1 within the cell contributes to the pathogenicity of avian influenza viruses.


Subject(s)
Influenza A virus/metabolism , Influenza A virus/pathogenicity , Influenza in Birds/virology , Interferon-beta/antagonists & inhibitors , Intracellular Space/virology , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Cells, Cultured , Chick Embryo , Chickens , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Influenza A virus/genetics , Influenza A virus/immunology , Influenza in Birds/immunology , Influenza in Birds/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Intracellular Space/immunology , Intracellular Space/metabolism , Protein Transport , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL