Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Cell Rep ; 35(2): 108966, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852868

ABSTRACT

Persistent virus infections can cause pathogenesis that is debilitating or lethal. During these infections, virus-specific T cells fail to protect due to weakened antiviral activity or failure to persist. These outcomes are governed by histone modifications, although it is unknown which enzymes contribute to T cell loss or impaired function over time. In this study, we show that T cell receptor-stimulated CD8+ T cells increase their expression of UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome) to enhance gene expression. During chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, UTX binds to enhancers and transcription start sites of effector genes, allowing for improved cytotoxic T lymphocyte (CTL)-mediated protection, independent of its trimethylation of histone 3 lysine 27 (H3K27me3) demethylase activity. UTX also limits the frequency and durability of virus-specific CD8+ T cells, which correspond to increased expression of inhibitory receptors. Thus, UTX guides gene expression patterns in CD8+ T cells, advancing early antiviral defenses while reducing the longevity of CD8+ T cell responses.


Subject(s)
Cytotoxicity, Immunologic/genetics , Histone Demethylases/genetics , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Gene Expression Profiling , Gene Expression Regulation , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Histone Demethylases/deficiency , Histone Demethylases/immunology , Histones/genetics , Histones/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes, Cytotoxic/virology , Viral Load/genetics , Viral Load/immunology , Lymphocyte Activation Gene 3 Protein
2.
Viruses ; 12(8)2020 08 16.
Article in English | MEDLINE | ID: mdl-32824338

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is a prototypic arenavirus. The function of untranslated regions (UTRs) of the LCMV genome has not been well studied except for the extreme 19 nucleotide residues of both the 5' and 3' termini. There are internal UTRs composed of 58 and 41 nucleotide residues in the 5' and 3' UTRs, respectively, in the LCMV S segment. Their functional roles have yet to be elucidated. In this study, reverse genetics and minigenome systems were established for LCMV strain WE and the function of these regions were analyzed. It was revealed that nucleotides 20-40 and 20-38 located downstream of the 19 nucleotides in the 5' and 3' termini, respectively, were involved in viral genome replication and transcription. Furthermore, it was revealed that the other internal UTRs (nucleotides 41-77 and 39-60 in the 5' and 3' termini, respectively) in the S segment were involved in virulence in vivo, even though these regions did not affect viral growth capacity in Vero cells. The introduction of LCMV with mutations in these regions attenuates the virus and may enable the production of LCMV vaccine candidates.


Subject(s)
Genome, Viral , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/genetics , Untranslated Regions/physiology , A549 Cells , Animals , Chlorocebus aethiops , Female , Humans , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mutation , RNA, Viral/chemistry , Reverse Genetics , Specific Pathogen-Free Organisms , Vero Cells , Virulence , Virus Replication
3.
J Virol ; 92(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29593035

ABSTRACT

Mammarenaviruses cause chronic infections in their natural rodent hosts. Infected rodents shed infectious virus into excreta. Humans are infected through mucosal exposure to aerosols or direct contact of abraded skin with fomites, resulting in a wide range of manifestations from asymptomatic or mild febrile illness to severe life-threatening hemorrhagic fever. The mammarenavirus matrix Z protein has been shown to be a main driving force of virus budding and to act as a negative regulator of viral RNA synthesis. To gain a better understanding of how the Z protein exerts its several different functions, we investigated the interaction between Z and viral polymerase L protein using the prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV). We found that in the presence of an active viral ribonucleoprotein (vRNP), the Z protein translocated from nonionic detergent-resistant, membrane-rich structures to a subcellular compartment with a different membrane composition susceptible to disruption by nonionic detergents. Alanine (A) substitution of a highly conserved leucine (L) at position 72 in LCMV Z protein abrogated Z-L interaction. The L72A mutation did not affect the stability or budding activity of Z when expressed alone, but in the presence of an active vRNP, mutation L72A promoted rapid degradation of Z via a proteasome- and lysosome-independent pathway. Accordingly, L72A mutation in the Z protein resulted in nonviable LCMV. Our findings have uncovered novel aspects of the dynamics of the Z protein for which a highly conserved L residue was strictly required.IMPORTANCE Several mammarenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever disease in humans and pose important public health concerns in their regions of endemicity. Moreover, mounting evidence indicates that the worldwide-distributed, prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), is a neglected human pathogen of clinical significance. The mammarenavirus matrix Z protein plays critical roles in different steps of the viral life cycle by interacting with viral and host cellular components. Here we report that alanine substitution of a highly conserved leucine residue, located at position 72 in LCMV Z protein, abrogated Z-L interaction. The L72A mutation did not affect Z budding activity but promoted its rapid degradation in the presence of an active viral ribonucleoprotein (vRNP). Our findings have uncovered novel aspects of the dynamics of the Z protein for which a highly conserved L residue was strictly required.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Lymphocytic choriomeningitis virus/growth & development , RNA-Dependent RNA Polymerase/metabolism , Ribonucleoproteins/metabolism , Viral Proteins/metabolism , Virus Release/genetics , Alanine/genetics , Amino Acid Substitution/genetics , Ammonium Chloride/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Leucine/genetics , Leupeptins/pharmacology , Lymphocytic choriomeningitis virus/genetics , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , RNA-Dependent RNA Polymerase/genetics , Vero Cells , Viral Proteins/genetics
4.
Immunity ; 47(6): 1129-1141.e5, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246443

ABSTRACT

During chronic stimulation, CD8+ T cells acquire an exhausted phenotype characterized by expression of inhibitory receptors, down-modulation of effector function, and metabolic impairments. T cell exhaustion protects from excessive immunopathology but limits clearance of virus-infected or tumor cells. We transcriptionally profiled antigen-specific T cells from mice infected with lymphocytic choriomeningitis virus strains that cause acute or chronic disease. T cell exhaustion during chronic infection was driven by high amounts of T cell receptor (TCR)-induced transcription factors IRF4, BATF, and NFATc1. These regulators promoted expression of inhibitory receptors, including PD-1, and mediated impaired cellular metabolism. Furthermore, they repressed the expression of TCF1, a transcription factor required for memory T cell differentiation. Reducing IRF4 expression restored the functional and metabolic properties of antigen-specific T cells and promoted memory-like T cell development. These findings indicate that IRF4 functions as a central node in a TCR-responsive transcriptional circuit that establishes and sustains T cell exhaustion during chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon Regulatory Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Gene Expression Regulation , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Signal Transduction
5.
J Virol Methods ; 249: 194-196, 2017 11.
Article in English | MEDLINE | ID: mdl-28923314

ABSTRACT

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.


Subject(s)
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
6.
Nature ; 537(7620): 412-428, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27501245

ABSTRACT

During chronic viral infection, virus-specific CD8(+) T cells become exhausted, exhibit poor effector function and lose memory potential. However, exhausted CD8(+) T cells can still contain viral replication in chronic infections, although the mechanism of this containment is largely unknown. Here we show that a subset of exhausted CD8(+) T cells expressing the chemokine receptor CXCR5 has a critical role in the control of viral replication in mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). These CXCR5(+) CD8(+) T cells were able to migrate into B-cell follicles, expressed lower levels of inhibitory receptors and exhibited more potent cytotoxicity than the CXCR5(-) [corrected] subset. Furthermore, we identified the Id2-E2A signalling axis as an important regulator of the generation of this subset. In patients with HIV, we also identified a virus-specific CXCR5(+) CD8(+) T-cell subset, and its number was inversely correlated with viral load. The CXCR5(+) subset showed greater therapeutic potential than the CXCR5(-) [corrected] subset when adoptively transferred to chronically infected mice, and exhibited synergistic reduction of viral load when combined with anti-PD-L1 treatment. This study defines a unique subset of exhausted CD8(+) T cells that has a pivotal role in the control of viral replication during chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Germinal Center/cytology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Receptors, CXCR5/metabolism , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Chronic Disease , Female , Germinal Center/immunology , HIV Infections/immunology , HIV Infections/virology , Humans , Inhibitor of Differentiation Protein 2/metabolism , Lymphocytic choriomeningitis virus/growth & development , Male , Mice , Receptors, CXCR5/deficiency , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Viral Load/immunology , Virus Replication/immunology
7.
J Virol ; 89(7): 3523-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25589652

ABSTRACT

UNLABELLED: Arenaviruses have a significant impact on public health and pose a credible biodefense threat, but the development of safe and effective arenavirus vaccines has remained elusive, and currently, no Food and Drug Administration (FDA)-licensed arenavirus vaccines are available. Here, we explored the use of a codon deoptimization (CD)-based approach as a novel strategy to develop live-attenuated arenavirus vaccines. We recoded the nucleoprotein (NP) of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with the least frequently used codons in mammalian cells, which caused lower LCMV NP expression levels in transfected cells that correlated with decreased NP activity in cell-based functional assays. We used reverse-genetics approaches to rescue a battery of recombinant LCMVs (rLCMVs) encoding CD NPs (rLCMV/NP(CD)) that showed attenuated growth kinetics in vitro. Moreover, experiments using the well-characterized mouse model of LCMV infection revealed that rLCMV/NP(CD1) and rLCMV/NP(CD2) were highly attenuated in vivo but, upon a single immunization, conferred complete protection against a subsequent lethal challenge with wild-type (WT) recombinant LCMV (rLCMV/WT). Both rLCMV/NP(CD1) and rLCMV/NP(CD2) were genetically and phenotypically stable during serial passages in FDA vaccine-approved Vero cells. These results provide proof of concept of the safety, efficacy, and stability of a CD-based approach for developing live-attenuated vaccine candidates against human-pathogenic arenaviruses. IMPORTANCE: Several arenaviruses cause severe hemorrhagic fever in humans and pose a credible bioterrorism threat. Currently, no FDA-licensed vaccines are available to combat arenavirus infections, while antiarenaviral therapy is limited to the off-label use of ribavirin, which is only partially effective and is associated with side effects. Here, we describe the generation of recombinant versions of the prototypic arenavirus LCMV encoding codon-deoptimized viral nucleoproteins (rLCMV/NP(CD)). We identified rLCMV/NP(CD1) and rLCMV/NP(CD2) to be highly attenuated in vivo but able to confer protection against a subsequent lethal challenge with wild-type LCMV. These viruses displayed an attenuated phenotype during serial amplification passages in cultured cells. Our findings support the use of this approach for the development of safe, stable, and protective live-attenuated arenavirus vaccines.


Subject(s)
Arenaviridae Infections/prevention & control , Codon , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/immunology , Viral Vaccines/immunology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Chlorocebus aethiops , Disease Models, Animal , Gene Expression , Genomic Instability , Lymphocytic choriomeningitis virus/genetics , Male , Mice , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/isolation & purification , Virus Cultivation , Virus Replication
8.
J Virol ; 88(15): 8340-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24829355

ABSTRACT

UNLABELLED: A key characteristic of arenaviruses is their ability to establish persistent infection in their natural host. Different factors like host age, viral dose strain, and route of infection may contribute to the establishment of persistence. However, the molecular mechanisms governing persistence are not fully understood. Here, we describe gain-of-function mutations of lymphocytic choriomeningitis virus (LCMV) expressing Lassa virus (LASV) GP, which can prolong viremia in mice depending on the sequences in the GP-2 cytoplasmic tail. The initial mutant variant (rLCMV/LASV mut GP) carried a point mutation in the cytosolic tail of the LASV glycoprotein GP corresponding to a K461G substitution. Unlike what occurred with the original rLCMV/LASV wild-type (wt) GP, infection of C57BL/6 mice with the mutated recombinant virus led to a detectable viremia of 2 weeks' duration. Further replacement of the entire sequence of the cytosolic tail from LASV to LCMV GP resulted in increased viral titers and delayed clearance of the viruses. Biosynthesis and cell surface localization of LASV wt and mut GPs were comparable. IMPORTANCE: Starting from an emerging virus in a wild-type mouse, we engineered a panel of chimeric Lassa/lymphocytic choriomeningitis viruses. Mutants carrying a viral envelope with the cytosolic tail from the closely related mouse-adapted LCMV were able to achieve a productive viral infection lasting up to 27 days in wild-type mice. Biochemical assays showed a comparable biosynthesis and cell surface localization of LASV wt and mut GPs. These recombinant chimeric viruses could allow the study of immune responses and antivirals targeting the LASV GP.


Subject(s)
Evolution, Molecular , Lassa virus/growth & development , Lassa virus/genetics , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/genetics , Recombination, Genetic , Animals , Antigens, Viral/genetics , Glycoproteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mutant Proteins/genetics , Mutant Proteins/metabolism , Point Mutation , Protein Structure, Tertiary/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Load , Viral Proteins/genetics , Viremia
9.
PLoS One ; 8(1): e53273, 2013.
Article in English | MEDLINE | ID: mdl-23308183

ABSTRACT

The glycoprotein (GP) of arenaviruses is glycosylated at 11 conserved N-glycosylation sites. We constructed recombinant lymphocytic choriomeningitis virus (rLCMV) featuring either additions or deletions of these N-glycans to investigate their role in the viral life cycle. N-glycosylation at two sites, T87 and S97, were found to be necessary to rescue rLCMV. Three of nine successfully rescued mutants, S116A, T234A, and S373A, under selective pressures in either epithelial, neuronal, or macrophage cells reverted to WT sequence. Of the seven stable N-glycan deletion mutants, five of these led to altered viral fitness and cell tropism, assessed as growth in either mouse primary cortical neurons or bone marrow derived macrophages. These results demonstrate that the deletion of N-glycans in LCMV GP may confer an advantage to the virus for infection of neurons but a disadvantage in macrophages.


Subject(s)
Glycoproteins/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Cells, Cultured , Glycoproteins/genetics , Glycosylation , Humans , Lymphocytic Choriomeningitis/veterinary , Lymphocytic choriomeningitis virus/genetics , Macrophages/virology , Mice , Mice, Inbred C57BL , Models, Molecular , Mutation , Neurons/virology , Polysaccharides/genetics , Polysaccharides/metabolism , Tropism , Viral Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 109(35): 14116-21, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22893686

ABSTRACT

Interleukin-10 (IL-10) is an important factor involved in T-cell dysfunction during persistent viral infection. Although several factors can negatively regulate T-cell activity, targeting of the IL-10 pathway alone is sufficient to regenerate T-cell activity and increase viral control. How IL-10 mediates these effects is unclear. Here, we investigated the cellular source of IL-10 necessary for establishing T-cell exhaustion and viral persistence, using IL-10 reporter mice (VertX), cell-type-specific IL-10 and IL-10 receptor deletion mice, and bone marrow chimeric mice. During establishment of viral persistence, the cellular subset with the most prevalent expression of IL-10 was CD8α(-)CD4(+) dendritic cells (DCs), which produced IL-10 with increasing kinetics until 9 d postinfection. After this time point, DCs exhibited a modest decline in percentage of IL-10(+) cells whereas B cells and CD4(+) T cells increased minimally. Further analysis of the DC population demonstrated that IL-10 was primarily expressed in infected DCs. These DCs were a notable source of IL-10 as mutant mice with a DC-specific deletion of IL-10 had significantly decreased serum levels. Interestingly, viral infection was not directly causative of IL-10 expression; rather, IL-10 production appeared to be linked to type I IFN signaling. Our findings further illuminate the contribution of DCs to the production of IL-10 and to viral persistence.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Interleukin-10/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/metabolism , Dendritic Cells/metabolism , Female , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Transplantation Chimera
11.
PLoS One ; 7(5): e37337, 2012.
Article in English | MEDLINE | ID: mdl-22615984

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV), a natural murine pathogen, is a member of the Arenavirus family, may cause atypical meningitis in humans, and has been utilized extensively as a model pathogen for the study of virus-induced disease and immune responses. Historically, viral titers have been quantified by a standard plaque assay, but for non-cytopathic viruses including LCMV this requires lengthy incubation, so results cannot be obtained rapidly. Additionally, due to specific technical constraints of the plaque assay including the visual detection format, it has an element of subjectivity along with limited sensitivity. In this study, we describe the development of a FACS-based assay that utilizes detection of LCMV nucleoprotein (NP) expression in infected cells to determine viral titers, and that exhibits several advantages over the standard plaque assay. We show that the LCMV-NP FACS assay is an objective and reproducible detection method that requires smaller sample volumes, exhibits a ∼20-fold increase in sensitivity to and produces results three times faster than the plaque assay. Importantly, when applied to models of acute and chronic LCMV infection, the LCMV-NP FACS assay revealed the presence of infectious virus in samples that were determined to be negative by plaque assay. Therefore, this technique represents an accelerated, enhanced and objective alternative method for detection of infectious LCMV that is amenable to adaptation for other viral infections as well as high throughput diagnostic platforms.


Subject(s)
Flow Cytometry/methods , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Nucleoproteins/biosynthesis , Viral Load/methods , Animals , Antibodies, Viral/biosynthesis , Lymphocytic Choriomeningitis/blood , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/metabolism , Mice , Nucleoproteins/immunology , Sensitivity and Specificity , Viral Plaque Assay , Viremia/diagnosis
12.
J Virol ; 85(24): 13069-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21957293

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Lymphocytic choriomeningitis virus/drug effects , Lymphocytic choriomeningitis virus/growth & development , Cell Hypoxia , HeLa Cells , Humans
13.
Cell Host Microbe ; 9(3): 212-222, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21402360

ABSTRACT

The prototypic arenavirus lymphocytic choriomeningitis virus (LCMV), which naturally persists in rodents, represents a model for HIV, HBV, and HCV. Cleavage of the viral glycoprotein precursor by membrane-bound transcription factor peptidase, site 1 (Mbtps1 or site-1 protease), is crucial for the life cycle of arenaviruses and therefore represents a potential target for therapy. Recently, we reported a viable hypomorphic allele of Mbtps1 (woodrat) encoding a protease with diminished enzymatic activity. Using the woodrat allele, we examine the role of Mbtps1 during persistent LCMV infection. Surprisingly, Mbtps1 inhibition limits persistent but not acute viral infection and is associated with an organ/cell type-specific decrease in viral titers. Analysis of bone marrow-derived dendritic cells from woodrat mice supports their specific role in resolving persistent viral infection. These results support in vivo targeting of Mbtps1 in the treatment of arenavirus infections and demonstrate a critical role for dendritic cells in persistent viral infections.


Subject(s)
Arenaviridae Infections/immunology , Lymphocytic choriomeningitis virus/physiology , Proprotein Convertases/immunology , Serine Endopeptidases/immunology , Animals , Animals, Genetically Modified/immunology , Animals, Genetically Modified/virology , Antigens, Viral/metabolism , Arenaviridae Infections/virology , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Dendritic Cells/immunology , Immunity, Innate , Immunocompromised Host , Kidney/immunology , Kidney/virology , Liver/immunology , Liver/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Mutation , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Spleen/immunology , Spleen/virology , T-Lymphocytes/immunology , Viral Plaque Assay , Viral Tropism
14.
Curr Protoc Microbiol ; Chapter 15: Unit 15A.1, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18770534

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is an enveloped, ambisense RNA virus and the prototypic virus of the arenavirus group. It can cause viral meningitis and other ailments in humans, but its natural host is the mouse. The LCMV/mouse model has been useful for examining mechanisms of viral persistence and the basic concepts of virus-induced immunity and immunopathology. This unit discusses strain differences and biosafety containment issues for LCMV. Recommendations are made for techniques for propagating LCMV to high titers to quantify it by plaque assay and PCR techniques and to preserve its infectivity by appropriate storage.


Subject(s)
Culture Techniques , Lymphocytic choriomeningitis virus/growth & development , Reverse Transcriptase Polymerase Chain Reaction/methods , Viral Plaque Assay/methods , Virology/methods , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Cryopreservation/methods , Occupational Health , RNA, Viral/genetics , RNA, Viral/isolation & purification , Vero Cells , Virus Cultivation
15.
Virology ; 379(2): 205-12, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18691729

ABSTRACT

Multiple viruses induce reactive oxygen intermediate (ROI) generation during infection that plays an important role in growth. We have examined the importance of ROI during lymphocytic choriomeningitis virus (LCMV) infection of immortalized BHK-21 cells and murine peritoneal macrophages. Within 15 min of virus addition, intracellular ROI levels increased. To examine the contribution of ROI to LCMV infection, cells were pretreated with antioxidant prior to virus addition. Antioxidant treatment inhibited low and high MOI growth of virus. The requirement for ROI was greatest during the initial phase of infection, as antioxidant treatment after 6 h post infection had a weaker inhibitory effect. Furthermore, antioxidant treatment of cells inhibited virus binding, while treatment of virus stocks with N-ethyl malemide, which blocks free thiols, eliminated infectious virus. This illustrates that ROI are critical to the regulation of virus binding and growth and has important implications for understanding the infectivity of related viruses.


Subject(s)
Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/physiology , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Azoles/pharmacology , Cell Line , Cricetinae , Ethylmaleimide/pharmacology , Isoindoles , Lymphocytic choriomeningitis virus/drug effects , Lymphocytic choriomeningitis virus/pathogenicity , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Metalloporphyrins/pharmacology , Mice , Organoselenium Compounds/pharmacology , Sulfhydryl Reagents/pharmacology , Virulence/drug effects , Virulence/physiology
16.
J Immunol ; 180(5): 3113-21, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292534

ABSTRACT

CD8+ T cells are crucial for the control of intracellular pathogens such as viruses and some bacteria. Using lymphocytic choriomeningitis virus (LCMV) infection of mice--the prototypic arenavirus evolutionarily closely related to human Lassa fever and South American hemorrhagic fever viruses, we have shown previously that the kinetics of Ag presentation determine immunodominance of the LCMV-specific CTL response due to progressive exhaustion of LCMV nucleoprotein (NP)-specific CTL upon increasing viral load. In this study, we provide evidence that CTL against early LCMV NP-derived epitopes are more important in virus control than those against late glycoprotein-derived epitopes. We show that mice that are tolerant to all NP-derived T cell epitopes are severely compromised in their ability to control larger inocula of LCMV, supporting our hypothesis that CD8+ T cells specific for early viral Ags play a major role in acute virus control. Thus, the kinetics with which virus-derived T cell epitopes are presented has a strong impact on the efficacy of the antiviral immunity. This aspect should be taken into consideration for the development of vaccines.


Subject(s)
Antigens, Viral/immunology , Cytotoxicity, Immunologic , Nucleoproteins/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/immunology , Immune Tolerance/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/metabolism , Viral Load
17.
Nat Med ; 13(11): 1316-23, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17982463

ABSTRACT

T helper cells can support the functions of CD8(+) T cells against persistently infecting viruses such as murine lymphocytic choriomeningitis virus (LCMV), cytomegalovirus, hepatitis C virus and HIV. These viruses often resist complete elimination and remain detectable at sanctuary sites, such as the kidneys and other extralymphatic organs. The mechanisms underlying this persistence are not well understood. Here we show that mice with potent virus-specific T-cell responses have reduced levels and delayed formation of neutralizing antibodies, and these mice fail to clear LCMV from extralymphatic epithelia. Transfer of virus-specific B cells but not virus-specific T cells augmented virus clearance from persistent sites. Virus elimination from the kidneys was associated with the formation of IgG deposits in the interstitial space, presumably from kidney-infiltrating B cells. CD8(+) T cells in the kidneys of mice that did not clear virus from this site were activated but showed evidence of exhaustion. Thus, we conclude that in this model of infection, site-specific virus persistence develops as a consequence of potent immune activation coupled with reductions in virus-specific neutralizing antibodies. Our results suggest that sanctuary-site formation depends both on organ anatomy and on the induction of different adaptive immune effector mechanisms. Boosting T-cell responses alone may not reduce virus persistence.


Subject(s)
Lymphatic System/immunology , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes/immunology , Virus Latency/immunology , Virus Replication/immunology , Animals , Cell Line , Cricetinae , Kidney/immunology , Kidney/virology , Liver/immunology , Liver/virology , Lung/immunology , Lung/virology , Lymphatic System/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , T-Lymphocytes/virology
18.
Life Sci ; 80(24-25): 2330-3, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17286988

ABSTRACT

A number of studies have demonstrated that non-neuronal acetylcholine can play a role in the regulation of T cell function. Recently, we reported that CD8(+) T cells, from mice with a targeted deletion of the M(1) muscarinic receptor, had a defect in differentiating into cytolytic T lymphocytes when stimulated in vitro. In the current report, we analyze the in vivo function of CD8(+) T cells from mice with targeted deletions of either M(1) or M(5) muscarinic receptors. M(1) or M(5) knockout mice were infected with either lymphocytic choriomeningitis virus or vesicular stomatitis virus. Expansion of anti-viral CD8(+) T cells was monitored by staining with tetramer reagents specific for the immunodominant peptides of the viruses. No defect in expansion of CD8(+) T cells was observed in either M(1) or M(5) knockout mice. The extent to which one can draw a generalized conclusion that M(1) and M(5) are not involved in anti-viral immunity depends upon issues of antigen strength, genetic background, induction of redundant receptors, and the potential for qualitative defects in the expanded CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Receptor, Muscarinic M1/physiology , Receptor, Muscarinic M5/physiology , Virus Diseases/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Female , Flow Cytometry , Gene Deletion , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M5/genetics , Time Factors , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/immunology
19.
Vaccine ; 25(12): 2279-87, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17258845

ABSTRACT

In the midst of new investigations into the mechanisms of both delivery and protection of new vaccines and vaccine carriers, it has become clear that immunization with delivery mechanisms that do not involve living, replicating organisms are vastly preferred. In this report, non-replicating bacterial minicells simultaneously co-delivering the nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) and the corresponding DNA vaccine were tested for the ability to generate protective cellular immune responses in mice. It was found that good protection (89%) was achieved after intramuscular administration, moderate protection (31%) was achieved after intranasal administration, and less protection (7%) was achieved following gastric immunization. These results provide a solid foundation on which to pursue the use of bacterial minicells as a non-replicating vaccine delivery platform.


Subject(s)
Immunization/methods , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Nucleoproteins/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Administration, Intranasal , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COS Cells , Chlorocebus aethiops , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Drug Delivery Systems/methods , Escherichia coli/virology , Injections, Intramuscular , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Nucleoproteins/genetics , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
20.
J Virol ; 78(20): 11443-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452271

ABSTRACT

The arenavirus small RING finger Z protein is the main driving force of arenavirus budding. The primary structure of Z is devoid of hydrophobic transmembrane domains, but both lymphocytic choriomeningitis virus (LCMV) and Lassa fever virus Z proteins accumulate near the inner surface of the plasma membrane and are strongly membrane associated. All known arenavirus Z proteins contain a glycine (G) at position 2, which is a potential acceptor site for a myristoyl moiety. Metabolic labeling showed incorporation of [(3)H]myristic acid by wild-type Z protein but not by the G2A mutant. The mutation G2A eliminated Z-mediated budding. Likewise, treatment with the myristoylation inhibitor 2-hydroxymyristic acid inhibited Z-mediated budding, eliminated formation of virus-like particles, and caused a dramatic reduction in virus production in LCMV-infected cells. Budding activity was restored in G2A mutant Z proteins by the addition of the myristoylation domain of the tyrosine protein kinase Src to their N termini. These findings indicate N-terminal myristoylation of Z plays a key role in arenavirus budding.


Subject(s)
Carrier Proteins/metabolism , Lassa virus/growth & development , Lymphocytic choriomeningitis virus/growth & development , Myristic Acid/metabolism , Viral Matrix Proteins/metabolism , Zinc Fingers/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Intracellular Signaling Peptides and Proteins , Lassa virus/metabolism , Lymphocytic choriomeningitis virus/metabolism , Mutation , RNA-Binding Proteins , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...