Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Virol J ; 20(1): 108, 2023 05 31.
Article En | MEDLINE | ID: mdl-37259080

BACKGROUND: Increased glucose uptake and utilization via aerobic glycolysis are among the most prominent hallmarks of tumor cell metabolism. Accumulating evidence suggests that similar metabolic changes are also triggered in many virus-infected cells. Viral propagation, like highly proliferative tumor cells, increases the demand for energy and macromolecular synthesis, leading to high bioenergetic and biosynthetic requirements. Although significant progress has been made in understanding the metabolic changes induced by viruses, the interaction between host cell metabolism and arenavirus infection remains unclear. Our study sheds light on these processes during lymphocytic choriomeningitis virus (LCMV) infection, a model representative of the Arenaviridae family. METHODS: The impact of LCMV on glucose metabolism in MRC-5 cells was studied using reverse transcription-quantitative PCR and biochemical assays. A focus-forming assay and western blot analysis were used to determine the effects of glucose deficiency and glycolysis inhibition on the production of infectious LCMV particles. RESULTS: Despite changes in the expression of glucose transporters and glycolytic enzymes, LCMV infection did not result in increased glucose uptake or lactate excretion. Accordingly, depriving LCMV-infected cells of extracellular glucose or inhibiting lactate production had no impact on viral propagation. However, treatment with the commonly used glycolytic inhibitor 2-deoxy-D-glucose (2-DG) profoundly reduced the production of infectious LCMV particles. This effect of 2-DG was further shown to be the result of suppressed N-linked glycosylation of the viral glycoprotein. CONCLUSIONS: Although our results showed that the LCMV life cycle is not dependent on glucose supply or utilization, they did confirm the importance of N-glycosylation of LCMV GP-C. 2-DG potently reduces LCMV propagation not by disrupting glycolytic flux but by inhibiting N-linked protein glycosylation. These findings highlight the potential for developing new, targeted antiviral therapies that could be relevant to a wider range of arenaviruses.


Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus , Humans , Glycosylation , Glycoproteins , Deoxyglucose/pharmacology
2.
Viruses ; 14(9)2022 08 27.
Article En | MEDLINE | ID: mdl-36146703

Over the millions of years-long co-evolution with their hosts, viruses have evolved plenty of mechanisms through which they are able to escape cellular anti-viral defenses and utilize cellular pathways and organelles for replication and production of infectious virions. In recent years, it has become clear that lipids play an important role during viral replication. Viruses use cellular lipids in a variety of ways throughout their life cycle. They not only physically interact with cellular membranes but also alter cellular lipid metabolic pathways and lipid composition to create an optimal replication environment. This review focuses on examples of how different viruses exploit cellular lipids in different cellular compartments during their life cycles.


Viruses , Cell Membrane , Lipids , Metabolic Networks and Pathways , Virus Replication
3.
Front Microbiol ; 10: 2438, 2019.
Article En | MEDLINE | ID: mdl-31708904

Experimental data indicate that during persistent infection, lymphocytic choriomeningitis virus (LCMV) may both directly or indirectly modulate regulatory cellular processes and alter cellular functions that are not critical for survival, but are essential for cell homeostasis. In order to shed more light on these processes, two-dimensional differential in-gel electrophoresis (2D-DIGE) and MALDI-TOF tandem mass spectrometry were used to determine the proteome response of the HeLa cell line to persistent LCMV infection. Quantitative analysis revealed 24 differentially abundant proteins. Functional analysis showed that LCMV-responsive proteins were primarily involved in metabolism, stress, and the defense response. Among identified proteins, we discovered significant changes for peroxiredoxins, a family of antioxidant enzymes. Decreased amount of these antioxidant proteins correlated with elevation of reactive oxygen species (ROS) in infected cells. Increased levels of ROS were accompanied by changes in the pattern of telomere restriction fragments (TRFs) in infected cells and mediated activation of hypoxia-inducible transcription factor-1 (HIF-1) and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways. Moreover, treatment with antioxidants resulted in reduced levels of viral nucleoprotein, indicating a connection between ROS-dependent signaling and viral replication.

4.
J Virol Methods ; 249: 194-196, 2017 11.
Article En | MEDLINE | ID: mdl-28923314

A common method for cell-associated virus isolation involves disruption of infected cells by a combination of hypotonic burst, freeze-thaw cycles (F-T) and sonication. This protocol was also originally used for the preparation of cell-free extract containing the MX strain of lymphocytic choriomeningitis virus (LCMV), which is preferentially propagated by cell-to-cell contact and does not release distinct virions into the medium. In the present study, we compared different approaches to virus isolation. Based on virus yield, we show that deionized water lysis is the fastest and most effective method for releasing LCMV MX infectious viral particles from persistently infected cells. Moreover, we demonstrate that freeze-thaw cycles and sonication do not improve virus isolation. This simple protocol could be used for isolation of other viruses, the life cycle of which is strictly cell-associated and therefore are difficult to release in large amounts from host cells.


Lymphocytic choriomeningitis virus/isolation & purification , Nucleoproteins/isolation & purification , Virion/isolation & purification , Virology/methods , A549 Cells , Animals , Antibodies, Viral , Buffers , Cell Culture Techniques , Chlorocebus aethiops , Culture Media/chemistry , Freezing , HeLa Cells , Humans , Lymphocytic Choriomeningitis/diagnosis , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Vero Cells
5.
J Interferon Cytokine Res ; 35(7): 540-53, 2015 Jul.
Article En | MEDLINE | ID: mdl-25830339

Lambda interferons inhibit replication of many viruses, but their role in the inhibition of lymphocytic choriomeningitis virus (LCMV) infection remains unclear. In this study, we examined the antiviral effects of interferon (IFN)-λ2 and IFN-λ3 against LCMV in A549 cells. We found that IFN-λ2 is a more potent inhibitor of LCMV strain MX compared with IFN-λ3, whereas both cytokines have similar antiviral effects against an immunosuppressive variant of LCMV, clone-13. We also demonstrated that the antiviral activity of IFN-λ2 is more effective if it is delivered early rather than after establishment of a long-term infection, suggesting that virus replication is only partially responsive to the cytokine. In agreement with this observation, we showed that LCMV infection significantly reduces IFNLR1 mRNA expression in infected cells. In addition, LCMV infection, to some extent, compromises the signal transduction pathway of IFN-λ2. This implies that IFN receptors as well as their downstream signaling components could be selectively targeted either directly by LCMV proteins or indirectly by cellular factor(s) that are induced or activated by LCMV infection.


Antiviral Agents/pharmacology , Interferons/pharmacology , Lymphocytic choriomeningitis virus/drug effects , Animals , Cell Line , Gene Expression Regulation/drug effects , Humans , Interferon-alpha/pharmacology , Kinetics , Lymphocytic Choriomeningitis/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interferon/metabolism , STAT1 Transcription Factor/metabolism , Transcription, Genetic/drug effects , Virus Replication/drug effects , Interferon gamma Receptor
6.
J Virol ; 85(24): 13069-76, 2011 Dec.
Article En | MEDLINE | ID: mdl-21957293

The physiological context of virus-infected cells can markedly affect multiplication and spread of the virus progeny. During persistent infection, the virus exploits the host cell without disturbing its vital functions. However, microenvironmental hypoxia can uncouple this intimate relationship and escalate virus pathogenesis. Accumulating evidence suggests that hypoxia-inducible factor (HIF) modulates gene expression of the viruses that pass through a DNA stage, contain hypoxia-responsive promoter elements, and replicate in the nucleus. Here we show that hypoxia can influence the gene expression and transmission of the cytoplasmic RNA virus lymphocytic choriomeningitis virus (LCMV), which is a neglected human pathogen and teratogen. The MX strain of LCMV, which we used as a model, replicates in a persistent mode in human HeLa cells, fails to produce mature envelope glycoproteins, and spreads through cell-cell contacts in the absence of extracellular infectious virions. Both exposure of MX-infected HeLa cells to chronic hypoxia and gene transfer approaches led to increased virus RNA transcription and higher levels of the viral proteins via a HIF-dependent mechanism. Moreover, hypoxia enhanced the formation of infectious virions capable of transmitting LCMV by cell-free medium. This LCMV "reactivation" might have health-compromising consequences in hypoxia-associated situations, such as fetal development and ischemia-related pathologies.


Gene Expression Regulation, Viral/drug effects , Lymphocytic choriomeningitis virus/drug effects , Lymphocytic choriomeningitis virus/growth & development , Cell Hypoxia , HeLa Cells , Humans
7.
J Virol ; 83(16): 7842-9, 2009 Aug.
Article En | MEDLINE | ID: mdl-19494018

Lymphocytic choriomeningitis virus (LCMV) is a prototypic arenavirus containing a bisegmented single-stranded RNA genome with an ambisense coding strategy. MX is a noncytolytic LCMV strain with an in vitro host range restricted to only few cell lines. MX LCMV spreads via cell-cell contacts and causes persistent infection with high production of viral nucleoprotein (NP). Using a proteomic approach, we identified keratin 1 (K1), an intermediate filament network component, as a binding partner of the viral NP. The functional significance of this interaction has been examined by chemical disruption of the keratin network, resulting in a reduced spread of MX LCMV in HeLa cells. However, K1 disassembly was considerably lower in MX LCMV-infected cells than in noninfected counterparts, indicating that NP can stabilize the keratin network and thereby support the integrity of cytoskeleton. The presence of NP also resulted in increased formation of desmosomes and stronger cell-cell adhesion. Similar effects were observed in HeLa cells persistently infected with LCMV strain Armstrong. Our findings suggest that the keratin network is important for the intercellular transmission of persistent LCMV infection in epithelial cells and show that the virus can actively facilitate its own intercellular spread through the interaction between the viral NP and K1 and stimulation of cell-cell contacts.


Keratin-1/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/physiology , Nucleoproteins/metabolism , Viral Proteins/metabolism , Cytoskeleton/metabolism , Cytoskeleton/virology , HeLa Cells , Humans , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Nucleoproteins/genetics , Protein Binding , Viral Proteins/genetics
8.
Toxicology ; 262(1): 73-9, 2009 Jul 28.
Article En | MEDLINE | ID: mdl-19460415

Calcium released from endoplasmic reticulum through special calcium release channels - inositol 1,4,5-trisphosphate receptors (IP(3)Rs) and ryanodine receptors (RyRs) - serves as a main source of cytosolic calcium signaling in the majority of cell types in physiological state and also in pathological situations. In this work, we studied whether IP(3)Rs can be involved in uranyl acetate induced nephrotoxicity. Using human embryonic kidney cell line (HEK293) as an experimental model we have found that uranyl acetate (5 and 50microM) up-regulates both, mRNA and protein levels of the type 1 and type 2 IP(3) receptors in HEK293 cells. This increase was associated with elevated expression of proapoptotic factors Bax and Caspase 3 and also by higher extent of apoptosis. Vice versa, induction of apoptosis resulted in increased mRNA levels of IP(3)R2 and also elevated levels of apoptotic markers. Therefore we propose that enhanced expression of the type 2 IP(3)Rs can at least partially contribute to increased levels of apoptosis due to uranyl acetate treatment.


Apoptosis/drug effects , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Organometallic Compounds/toxicity , Up-Regulation/drug effects , Calcium/metabolism , Caspase 3/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Dose-Response Relationship, Drug , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney/cytology , Kidney/drug effects , Organometallic Compounds/administration & dosage , RNA, Messenger/drug effects , RNA, Messenger/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
9.
Virus Genes ; 37(1): 31-8, 2008 Aug.
Article En | MEDLINE | ID: mdl-18493846

Lymphocytic choriomeningitis virus (LCMV) is the prototype Arenavirus with ambisense coding strategy. We have previously described a new MX strain LCMV and determined the primary structure of the genes coding for the nucleoprotein and RING finger Z protein. In this report, we describe amplification and sequencing of the entire coding sequences of additional MX genes, the glycoprotein precursor (GPC) and L protein. The obtained MX GPC cDNA sequence was 1,615 nucleotides long and contained an ORF, which encodes the GPC precursor of 498 amino acids. MX L polymerase cDNA sequence was 6,668 nucleotides long and predicted ORF encodes the L polymerase of 2,209 amino acids. Nucleotide and deduced amino acid sequences were compared with the known GPC and L sequences and the comparison revealed that both genes shared the highest amino acid identity with Armstrong strain. Phylogenetic analysis confirmed that MX represents a separate LCMV strain. The GPC and L genes products contained several characteristic conserved regions. On the other hand, we have observed numerous differences in predicted protein sequences, which distinguish MX LCMV from other LCMV strains and might be of potential biological significance.


Glycoproteins/genetics , Lymphocytic choriomeningitis virus/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Cell Line , Glycoproteins/chemistry , Glycoproteins/metabolism , HeLa Cells , Humans , Lymphocytic choriomeningitis virus/chemistry , Lymphocytic choriomeningitis virus/classification , Lymphocytic choriomeningitis virus/isolation & purification , Molecular Sequence Data , Open Reading Frames , Phylogeny , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Sequence Analysis, DNA , Viral Proteins/chemistry , Viral Proteins/metabolism
10.
Virus Res ; 132(1-2): 257-62, 2008 Mar.
Article En | MEDLINE | ID: mdl-18221814

Murid herpesvirus 4 (MuHV-4) is a member of the Gammaherpesvirus subfamily capable to establish a long-lasting latency and induce occasional malignancies. Because MuHV-4 is associated with cancer in a subset of virus-infected mice and because tumor development is often linked with hypoxia, we studied the influence of hypoxia on the biology of this virus. Using immunofluorescence and FACS analysis we detected increased proportion of MuHV-4 positive cells in the latently infected NB-78 cell line exposed to low oxygen conditions compared to normoxic controls. Moreover, the expression of ORF50, a crucial gene responsible for switch from latency to lytic virus replication, was induced upon the exposure of NB-78 cells to hypoxia. Luciferase reporter assays with ORF50 promoter confirmed the hypoxia-dependent induction. Transient co-transfections with hypoxia inducible factors showed that HIF-2alpha is a more potent activator of ORF50 expression than HIF-1alpha. Our results confirm that the MuHV-4 life cycle is influenced by low oxygen concentration.


Open Reading Frames , Oxygen/metabolism , Rhadinovirus/physiology , Transcription Factors/genetics , Up-Regulation , Virus Activation , Animals , Antigens, Viral/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Viral , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Oxygen/pharmacology , Promoter Regions, Genetic , Rhadinovirus/genetics , Time Factors , Transcription Factors/metabolism , Virus Latency
11.
Pflugers Arch ; 455(2): 349-57, 2007 Nov.
Article En | MEDLINE | ID: mdl-17611769

We studied the effects of the chloride channel blockers, 5-nitro-2-(phenylpropylamino)-benzoate (NPPB), dihydro-4,4' diisothiocyanostilbene-2,2'-disulphonic acid (DIDS), and phloretin on H2O2-induced primary culture cardiomyocyte apoptosis and activity of intracellular chloride channels obtained from rat heart mitochondrial and lysosomal vesicles. The chloride channel blockers (100 micromol/l) inhibited the H2O2-induced cardiomyocytes apoptosis. We characterized the effect of the blockers on single channel properties of the chloride channels derived from the mitochondrial and lysosomal vesicles incorporated into a bilayer lipid membrane. The single chloride channel currents were measured in 250:50 mmol/l KCl cis/trans solutions. NPPB, DIDS, and phloretin inhibited the chloride channels by decreasing the channel open probability in a concentration-dependent manner with EC50 values of 42, 7, and 20 micromol/l, respectively. NPPB and phloretin inhibited the channel's conductance and open dwell time, indicating that they could affect the chloride selective filter, pore permeability, and gating mechanism of the chloride channels. DIDS and NPPB inhibited the channels from the other side than bongkrekic acid and carboxyatractyloside. The results may contribute to understand a possible involvement of intracellular chloride channels in apoptosis and cardioprotection.


4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Chloride Channels/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Nitrobenzoates/pharmacology , Phloretin/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Chloride Channels/drug effects , Chloride Channels/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Hydrogen Peroxide/pharmacology , Lysosomes/metabolism , Male , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Rats , Rats, Wistar
...