Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
J Virol ; 98(3): e0188323, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38376197

RESUMO

Many viruses, including mammarenaviruses, have evolved mechanisms to counteract different components of the host cell innate immunity, which is required to facilitate robust virus multiplication. The double-stranded RNA (dsRNA) sensor protein kinase receptor (PKR) pathway plays a critical role in the cell anti-viral response. Whether PKR can restrict the multiplication of the Old World mammarenavirus lymphocytic choriomeningitis virus (LCMV) and the mechanisms by which LCMV may counteract the anti-viral functions of PKR have not yet been investigated. Here we present evidence that LCMV infection results in very limited levels of PKR activation, but LCMV multiplication is enhanced in the absence of PKR. In contrast, infection with a recombinant LCMV with a mutation affecting the 3'-5' exonuclease (ExoN) activity of the viral nucleoprotein resulted in robust PKR activation in the absence of detectable levels of dsRNA, which was associated with severely restricted virus multiplication that was alleviated in the absence of PKR. However, pharmacological inhibition of PKR activation resulted in reduced levels of LCMV multiplication. These findings uncovered a complex role of the PKR pathway in LCMV-infected cells involving both pro- and anti-viral activities.IMPORTANCEAs with many other viruses, the prototypic Old World mammarenavirus LCMV can interfere with the host cell innate immune response to infection, which includes the dsRNA sensor PKR pathway. A detailed understanding of LCMV-PKR interactions can provide novel insights about mammarenavirus-host cell interactions and facilitate the development of effective anti-viral strategies against human pathogenic mammarenaviruses. In the present work, we present evidence that LCMV multiplication is enhanced in PKR-deficient cells, but pharmacological inhibition of PKR activation unexpectedly resulted in severely restricted propagation of LCMV. Likewise, we document a robust PKR activation in LCMV-infected cells in the absence of detectable levels of dsRNA. Our findings have revealed a complex role of the PKR pathway during LCMV infection and uncovered the activation of PKR as a druggable target for the development of anti-viral drugs against human pathogenic mammarenaviruses.


Assuntos
Arenaviridae , Coriomeningite Linfocítica , Humanos , Arenaviridae/metabolismo , Linhagem Celular , Proteínas Quinases/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas de Transporte , Antivirais , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
2.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(9): 1296-1303, 2023.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38044640

RESUMO

OBJECTIVES: The differentiation of CD4+ T cells is regulated by a complex and fine signaling pathway composed of many molecules during immune response, and the molecular mechanism for regulating T-bet expression is unclear. Mediator complex subunit 1 (Med1) can combine with a variety of co-factors to regulate gene transcription, promote cell proliferation and survival, and affect invariant natural killer T cell (iNKT) development. This study aims to investigate the effect of Med1 on T cell development and CD4+ T cell differentiation in immune response. METHODS: Mice with T cell-specific knockout of Med1 gene (Med1F/FCD4cre+, KO) were constructed and verified. The percentage and number of CD4+ and CD8+ T cells in thymus, spleen, and lymph nodes of KO mice and control (Con) mice (Med1F/FCD4cre-) were detected by flow cytometry. After 8 days of infection with lymphocytic choriomeningitis virus (LCMV), the percentage and number of CD4+ T cells or antigen-specific (GP66+) CD4+ T cells, the percentage and number of Th1 cells (Ly6c+PSGL1+) in CD4+ T cells or antigen-specific CD4+ T cells were examined in the spleen of mice. Moreover, the fluorescence intensity of T-bet in CD4+ T cells or antigen-specific CD4+ T cells was analyzed. RESULTS: Compared with the Con group, the percentage and number of CD4+ T cells and CD8+ T cells in the thymus, CD4+ T cells in the spleen and lymph nodes of the KO group showed no significant differences (all P>0.05), but the percentage and number of CD8+ T cells in the spleen and lymph nodes of the KO group were diminished significantly (all P<0.05). After 8 days of infection with LCMV, there was no significant difference in the percentage and number of CD4+ T cells or antigen-specific CD4+ T cells in the spleen between the KO group and the Con group (all P>0.05), while in comparison with the Con group, the percentage and number of Th1 cells in CD4+ T cells or antigen-specific CD4+ T cells, and the expression of T-bet in CD4+ T cells or antigen-specific CD4+ T cells were significantly reduced in the spleen of the KO group (all P<0.05). CONCLUSIONS: Specific knockout of Med1 in T cells does not affect the development of CD4+ and CD8+ T cells in the thymus, but does affect the maintenance of peripheral CD8+ T cells. In the immune response, Med1 gene deletion affects the expression of transcription factor T-bet, which in turn to reduce Th1 cell differentiation.


Assuntos
Linfócitos T CD8-Positivos , Subunidade 1 do Complexo Mediador , Camundongos , Animais , Linfócitos T CD8-Positivos/metabolismo , Subunidade 1 do Complexo Mediador/metabolismo , Imunidade , Diferenciação Celular , Vírus da Coriomeningite Linfocítica/metabolismo , Células Th1/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Camundongos Endogâmicos C57BL
3.
Cell Death Dis ; 14(2): 123, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36792599

RESUMO

Necroptosis is a lytic and inflammatory form of cell death that is highly constrained to mitigate detrimental collateral tissue damage and impaired immunity. These constraints make it difficult to define the relevance of necroptosis in diseases such as chronic and persistent viral infections and within individual organ systems. The role of necroptotic signalling is further complicated because proteins essential to this pathway, such as receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL), have been implicated in roles outside of necroptotic signalling. We sought to address this issue by individually defining the role of RIPK3 and MLKL in chronic lymphocytic choriomeningitis virus (LCMV) infection. We investigated if necroptosis contributes to the death of LCMV-specific CD8+ T cells or virally infected target cells during infection. We provide evidence showing that necroptosis was redundant in the pathogenesis of acute forms of LCMV (Armstrong strain) and the early stages of chronic (Docile strain) LCMV infection in vivo. The number of immune cells, their specificity and reactivity towards viral antigens and viral loads are not altered in the absence of either MLKL or RIPK3 during acute and during the early stages of chronic LCMV infection. However, we identified that RIPK3 promotes immune dysfunction and prevents control of infection at later stages of chronic LCMV disease. This was not phenocopied by the loss of MLKL indicating that the phenotype was driven by a necroptosis-independent function of RIPK3. We provide evidence that RIPK3 signaling evoked a dysregulated type 1 interferone response which we linked to an impaired antiviral immune response and abrogated clearance of chronic LCMV infection.


Assuntos
Vírus da Coriomeningite Linfocítica , Proteínas Quinases , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Necroptose , Linfócitos T CD8-Positivos/metabolismo , Morte Celular , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
4.
PLoS Pathog ; 17(10): e1009996, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34648606

RESUMO

Members of the Old World Arenaviruses primarily utilize α-dystroglycan (α-DAG1) as a cellular receptor for infection. Mutations within the glycoprotein (GP) of lymphocytic choriomeningitis virus (LCMV) reduce or abrogate the binding affinity to α-DAG1 and thus influence viral persistence, kinetics, and cell tropism. The observation that α-DAG1 deficient cells are still highly susceptible to low affinity variants, suggests the use of an alternative receptor(s). In this study, we used a genome-wide CRISPR Cas9 knockout screen in DAG1 deficient 293T cells to identify host factors involved in α-DAG1-independent LCMV infection. By challenging cells with vesicular stomatitis virus (VSV), pseudotyped with the GP of LCMV WE HPI (VSV-GP), we identified the heparan sulfate (HS) biosynthesis pathway as an important host factor for low affinity LCMV infection. These results were confirmed by a genetic approach targeting EXTL3, a key factor in the HS biosynthesis pathway, as well as by enzymatic and chemical methods. Interestingly, a single point mutation within GP1 (S153F or Y155H) of WE HPI is sufficient for the switch from DAG1 to HS binding. Furthermore, we established a simple and reliable virus-binding assay, using directly labelled VSV-GP by intramolecular fusion of VSV-P and mWasabi, demonstrating the importance of HS for virus attachment but not entry in Burkitt lymphoma cells after reconstitution of HS expression. Collectively, our study highlights the essential role of HS for low affinity LCMV infection in contrast to their high affinity counterparts. Residual LCMV infection in double knockouts indicate the use of (a) still unknown entry receptor(s).


Assuntos
Proteoglicanas de Heparan Sulfato/metabolismo , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/transmissão , Vírus da Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/patogenicidade , Células HEK293 , Humanos , Receptores Virais/metabolismo
5.
Cell Rep ; 29(8): 2202-2216.e5, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31747595

RESUMO

Generation of protective immunity to infections and vaccinations declines with age. Studies in healthy individuals have implicated reduced miR-181a expression in T cells as contributing to this defect. To understand the impact of miR-181a expression on antiviral responses, we examined LCMV infection in mice with miR-181ab1-deficient T cells. We found that miR-181a deficiency delays viral clearance, thereby biasing the immune response in favor of CD4 over CD8 T cells. Antigen-specific CD4 T cells in mice with miR-181a-deficient T cells expand more and have a broader TCR repertoire with preferential expansion of high-affinity T cells than in wild-type mice. Importantly, generation of antigen-specific miR-181a-deficient CD8 effector T cells is particularly impaired, resulting in lower frequencies of CD8 T cells in the liver even at time points when the infection has been cleared. Consistent with the mouse model, CD4 memory T cells in individuals infected with West Nile virus at older ages tend to be more frequent and of higher affinity.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/fisiologia , Vírus da Coriomeningite Linfocítica/metabolismo , MicroRNAs/metabolismo , Envelhecimento/fisiologia , Animais , Modelos Animais de Doenças , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/genética , Camundongos , MicroRNAs/genética
6.
Int J Cancer ; 145(7): 1958-1969, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30972741

RESUMO

The efficacy of cancer vaccines has been limited by the immunosuppressive tumor microenvironment, which can be alleviated by immune checkpoint inhibitor (ICI) therapy. Here, we tested if oncolytic viruses (OVs), similar to ICI, can also synergize with cancer vaccines by modulating the tumor microenvironment. VSV-GP, a chimeric vesicular stomatitis virus (VSV) pseudotyped with the glycoprotein (GP) of the lymphocytic choriomeningitis virus, is a promising new OV candidate. Here, we show that in mouse B16-OVA melanoma, combination treatment of VSV-GP with an ovalbumin (OVA) peptide-loaded dendritic cell (DC) vaccine (DCVacc) significantly enhanced survival over the single agent therapies, although both DCVacc and DCVacc/VSV-GP treatments induced comparable levels of OVA-specific CD8 T cell responses. Virus replication was minimal so that direct viral oncolysis in B16-OVA did not contribute to this synergism. The strong therapeutic effect of the DCVacc/VSV-GP combination treatment was associated with high numbers of tumor-infiltrating, highly activated T cells and the relative reduction of regulatory T cells in treated and contra-lateral nontreated tumors. Accordingly, depletion of CD8 T cells but not natural killer cells abrogated the therapeutic effect of DCVacc/VSV-GP supporting the crucial role of CD8 T cells. In addition, a drastic increase in several proinflammatory cytokines was observed in VSV-GP-treated tumors. Taken together, OVs, similar to ICI, have the potential to markedly increase the efficacy of cancer vaccines by alleviating local immune suppression in the tumor microenvironment.


Assuntos
Vacinas Anticâncer/administração & dosagem , Glicoproteínas/metabolismo , Melanoma Experimental/terapia , Terapia Viral Oncolítica/métodos , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Vacinas Anticâncer/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Glicoproteínas/genética , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Melanoma Experimental/imunologia , Camundongos , Vírus Oncolíticos/fisiologia , Ovalbumina/imunologia , Resultado do Tratamento , Microambiente Tumoral/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
PLoS Pathog ; 14(2): e1006892, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29462184

RESUMO

Several mammalian arenaviruses (mammarenaviruses) cause hemorrhagic fevers in humans and pose serious public health concerns in their endemic regions. Additionally, mounting evidence indicates that the worldwide-distributed, prototypic mammarenavirus, lymphocytic choriomeningitis virus (LCMV), is a neglected human pathogen of clinical significance. Concerns about human-pathogenic mammarenaviruses are exacerbated by of the lack of licensed vaccines, and current anti-mammarenavirus therapy is limited to off-label use of ribavirin that is only partially effective. Detailed understanding of virus/host-cell interactions may facilitate the development of novel anti-mammarenavirus strategies by targeting components of the host-cell machinery that are required for efficient virus multiplication. Here we document the generation of a recombinant LCMV encoding a nucleoprotein (NP) containing an affinity tag (rLCMV/Strep-NP) and its use to capture the NP-interactome in infected cells. Our proteomic approach combined with genetics and pharmacological validation assays identified ATPase Na+/K+ transporting subunit alpha 1 (ATP1A1) and prohibitin (PHB) as pro-viral factors. Cell-based assays revealed that ATP1A1 and PHB are involved in different steps of the virus life cycle. Accordingly, we observed a synergistic inhibitory effect on LCMV multiplication with a combination of ATP1A1 and PHB inhibitors. We show that ATP1A1 inhibitors suppress multiplication of Lassa virus and Candid#1, a live-attenuated vaccine strain of Junín virus, suggesting that the requirement of ATP1A1 in virus multiplication is conserved among genetically distantly related mammarenaviruses. Our findings suggest that clinically approved inhibitors of ATP1A1, like digoxin, could be repurposed to treat infections by mammarenaviruses pathogenic for humans.


Assuntos
Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Nucleoproteínas/metabolismo , Mapas de Interação de Proteínas , Proteoma/análise , Proteínas Repressoras/fisiologia , ATPase Trocadora de Sódio-Potássio/fisiologia , Células A549 , Animais , Arenaviridae/fisiologia , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Proibitinas , Ligação Proteica , Proteínas Repressoras/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Células Vero
8.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237830

RESUMO

Fatal infection with the highly pathogenic Lassa virus (LASV) is characterized by extensive viral dissemination, indicating broad tissue tropism. The major cellular receptor for LASV is the highly conserved extracellular matrix receptor dystroglycan (DG). Binding of LASV depends on DG's tissue-specific posttranslational modification with the unusual O-linked polysaccharide matriglycan. Interestingly, functional glycosylation of DG does not always correlate with viral tropism observed in vivo The broadly expressed phosphatidylserine (PS) receptors Axl and Tyro3 were recently identified as alternative LASV receptor candidates. However, their role in LASV entry is not entirely understood. Here, we examine LASV receptor candidates in primary human cells and found coexpression of Axl with differentially glycosylated DG. To study LASV receptor use in the context of productive arenavirus infection, we employed recombinant lymphocytic choriomeningitis virus expressing LASV glycoprotein (rLCMV-LASV GP) as a validated biosafety level 2 (BSL2) model. We confirm and extend previous work showing that Axl can contribute to LASV entry in the absence of functional DG using "apoptotic mimicry" in a way similar to that of other enveloped viruses. We further show that Axl-dependent LASV entry requires receptor activation and involves a pathway resembling macropinocytosis. Axl-mediated LASV entry is facilitated by heparan sulfate and critically depends on the late endosomal protein LAMP-1 as an intracellular entry factor. In endothelial cells expressing low levels of functional DG, both receptors are engaged by the virus and can contribute to productive entry. In sum, we characterize the role of Axl in LASV entry and provide a rationale for targeting Axl in antiviral therapy.IMPORTANCE The highly pathogenic arenavirus Lassa virus (LASV) represents a serious public health problem in Africa. Although the principal LASV receptor, dystroglycan (DG), is ubiquitously expressed, virus binding critically depends on DG's posttranslational modification, which does not always correlate with tissue tropism. The broadly expressed phosphatidylserine receptor Axl was recently identified as an alternative LASV receptor candidate, but its role in LASV entry is unclear. Here, we investigate the exact role of Axl in LASV entry as a function of DG's posttranslational modification. We found that in the absence of functional DG, Axl can mediate LASV entry via apoptotic mimicry. Productive entry requires virus-induced receptor activation, involves macropinocytosis, and critically depends on LAMP-1. In endothelial cells that express low levels of glycosylated DG, both receptors can promote LASV entry. In sum, our study defines the roles of Axl in LASV entry and provides a rationale for targeting Axl in antiviral therapy.


Assuntos
Distroglicanas/metabolismo , Vírus Lassa/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Internalização do Vírus , Células A549 , Antivirais/farmacologia , Infecções por Arenaviridae/metabolismo , Linhagem Celular Tumoral , Distroglicanas/genética , Endossomos/metabolismo , Expressão Gênica , Glicosilação , Células HEK293 , Células HeLa , Heparitina Sulfato/farmacologia , Humanos , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/patogenicidade , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Pinocitose/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Receptores Proteína Tirosina Quinases/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Tropismo , Receptor Tirosina Quinase Axl
9.
Cell Rep ; 21(9): 2528-2540, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29186689

RESUMO

NK cells can reduce anti-viral T cell immunity during chronic viral infections, including infection with the lymphocytic choriomeningitis virus (LCMV). However, regulating factors that maintain the equilibrium between productive T cell and NK cell immunity are poorly understood. Here, we show that a large viral load resulted in inhibition of NK cell activation, which correlated with increased expression of Qa-1b, a ligand for inhibitory NK cell receptors. Qa-1b was predominantly upregulated on B cells following LCMV infection, and this upregulation was dependent on type I interferons. Absence of Qa-1b resulted in increased NK cell-mediated regulation of anti-viral T cells following viral infection. Consequently, anti-viral T cell immunity was reduced in Qa-1b- and NKG2A-deficient mice, resulting in increased viral replication and immunopathology. NK cell depletion restored anti-viral immunity and virus control in the absence of Qa-1b. Taken together, our findings indicate that lymphocytes limit NK cell activity during viral infection in order to promote anti-viral T cell immunity.


Assuntos
Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Animais , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo
10.
Exp Hematol ; 56: 7-15, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28911907

RESUMO

MicroRNA-22 (miR-22) is a highly conserved microRNA that can regulate cell proliferation, oncogenesis, and cell maturation, especially during stress. In hematopoietic stem cells (HSCs), miR-22 has been reported to be involved in the regulation of key self-renewal factors, including Tet2. Recent work demonstrates that miR-22 also participates in regulation of the interferon (IFN) response, and expression profiling studies suggest that it is variably expressed at different stages in erythroid differentiation. We thus hypothesized that miR-22 regulates maturation of erythroid progenitors during stress hematopoiesis through its interaction with IFN. We compared the blood and bone marrow of wild-type (WT) and miR-22-deficient mice at baseline and upon infectious challenge with systemic lymphochoriomeningitis (LCMV) virus. miR-22-deficient mice maintained platelet counts better than WT mice during infection, but they showed significantly reduced red blood cells and hemoglobin. Analysis of bone marrow progenitors demonstrated better overall survival and improved HSC homeostasis in infected miR-22-null mice compared with WT, which was attributable to a blunted IFN response to LCMV challenge in the miR-22-null mice. We found that miR-22 was expressed exclusively in stage II erythroid precursors and downregulated upon infection in WT mice. Our results indicate that miR-22 promotes the IFN response to viral infection and that it functions at baseline as a brake to slow erythroid differentiation and maintain adequate erythroid potential. Impaired regulation of erythrogenesis in the absence of miR-22 can lead to anemia during infection.


Assuntos
Anemia/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoese , Interferon-alfa/metabolismo , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , MicroRNAs/metabolismo , Estresse Fisiológico , Anemia/genética , Anemia/virologia , Animais , Células Precursoras Eritroides/patologia , Perfilação da Expressão Gênica , Interferon-alfa/genética , Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética
11.
Cancer Discov ; 6(10): OF3, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27554611

RESUMO

Two studies have illuminated some of the molecular underpinnings of T-cell exhaustion. The first pinpoints the subset of exhausted T cells that revive upon PD-1 blockade. The second describes key metabolic deficiencies-restricted glucose uptake and mitochondrial dysfunction-that drive T cells to exhaustion.


Assuntos
Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/metabolismo , Animais , Anticorpos/farmacologia , Proliferação de Células/efeitos dos fármacos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Humanos , Coriomeningite Linfocítica , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Receptores CXCR5/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
12.
Cell Physiol Biochem ; 38(4): 1343-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27007114

RESUMO

BACKGROUND: Graft versus host disease (GvHD) occurs in 20% of cases with patients having an MHC I matched bone marrow transplantation (BMT). Mechanisms causing this disease remain to be studied. METHODS: Here we used a CD8+ T cell transgenic mouse line (P14/CD45.1+) and transgenic DEE mice bearing ubiquitously the glycoprotein 33-41 (GP33) antigen derived from the major lymphocytic choriomeningitis virus (LCMV) epitope to study mechanisms of tolerance in anti-host reactive CD8+ T cells after BMT. RESULTS: We found that anti-host reactive CD8+ T cells (P14 T cells) were not negatively selected in the thymus and that they were present in wild type (WT) recipient mice as well as in DEE recipient mice. Anti-host reactive CD8+ T cells ignored the GP33 antigen expressed ubiquitously by host cells but they could be activated ex vivo via LCMV-infection. Lipopolysaccharides (LPS) induced transient cell damage in DEE mice bearing anti-host reactive CD8+ T cells after BMT, suggesting that induction of host inflammatory response could break antigen ignorance. Introducing the GP33 antigen into BM cells led to deletion of anti-host reactive CD8+ T cells. CONCLUSION: We found that after BMT anti-host reactive CD8+ T cells ignored host antigen in recipients and that they were only deleted when host antigen was present in hematopoietic cells. Moreover, LPS-induced immune activation contributed to induction of alloreactivity of anti-host reactive CD8+ T cells after BMT.


Assuntos
Transplante de Medula Óssea , Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica , Alanina Transaminase/metabolismo , Animais , Anticorpos/imunologia , Antígenos Virais/genética , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Epitopos/imunologia , Citometria de Fluxo , Glicoproteínas/genética , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/metabolismo , L-Lactato Desidrogenase/metabolismo , Lipopolissacarídeos/toxicidade , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transplante Homólogo , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
14.
Proc Natl Acad Sci U S A ; 111(24): 8925-30, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24889626

RESUMO

The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.


Assuntos
Interferon Tipo I/metabolismo , Febre Lassa/virologia , Doenças Vasculares/virologia , Animais , Lavagem Broncoalveolar , Linhagem Celular , Cricetinae , Citocinas/metabolismo , Feminino , Vírus Lassa , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Transgênicos , Transdução de Sinais , Células-Tronco/química , Linfócitos T Citotóxicos/virologia , Ativação Viral
15.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 6): 1764-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24914986

RESUMO

The X-ray crystal structure of the Lymphocytic choriomeningitis virus nucleoprotein C-terminal immunosuppressive domain (LCMV NPΔ340) was determined to 2.0 Šresolution. The structure indicates that LCMV NPΔ340, like the other structurally characterized arenaviral nucleoproteins, adopts the fold of an exonuclease. This structure provides a crucial three-dimensional template for functional exploration of the replication and immunosuppression of this prototypic arenavirus.


Assuntos
Tolerância Imunológica , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Vírus da Coriomeningite Linfocítica/fisiologia , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Proteínas Virais/química
16.
J Virol ; 88(11): 6243-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648461

RESUMO

UNLABELLED: Prime-boost immunization regimens have proven efficacious at generating robust immune responses. However, whether the level of replication of the boosting antigen impacts the magnitude and protective efficacy of vaccine-elicited immune responses remains unclear. To evaluate this, we primed mice with replication-defective adenovirus vectors expressing the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP), followed by boosting with either LCMV Armstrong, which is rapidly controlled, or LCMV CL-13, which leads to a more prolonged exposure to the boosting antigen. Although priming of naive mice with LCMV CL-13 normally results in T cell exhaustion and establishment of chronic infection, boosting with CL-13 resulted in potent recall CD8 T cell responses that were greater than those following boosting with LCMV Armstrong. Furthermore, following the CL-13 boost, a greater number of anamnestic CD8 T cells localized to the lymph nodes, exhibited granzyme B expression, and conferred improved protection against Listeria and vaccinia virus challenges compared with the Armstrong boost. Overall, our findings suggest that the replicative capacity of the boosting antigen influences the protective efficacy afforded by prime-boost vaccine regimens. These findings are relevant for optimizing vaccine candidates and suggest a benefit of robustly replicating vaccine vectors. IMPORTANCE: The development of optimal prime-boost vaccine regimens is a high priority for the vaccine development field. In this study, we compared two boosting antigens with different replicative capacities. Boosting with a more highly replicative vector resulted in augmented immune responses and improved protective efficacy.


Assuntos
Imunidade Heteróloga/imunologia , Imunização Secundária/métodos , Vacinas Virais/imunologia , Replicação Viral/fisiologia , Adenoviridae , Animais , Antígenos Heterófilos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Citometria de Fluxo , Vetores Genéticos , Glicoproteínas/metabolismo , Estimativa de Kaplan-Meier , Listeria/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estatísticas não Paramétricas , Vaccinia virus/imunologia
17.
PLoS Pathog ; 9(5): e1003395, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23737750

RESUMO

Viral infections of central nervous system (CNS) often trigger inflammatory responses that give rise to a wide range of pathological outcomes. The CNS is equipped with an elaborate network of innate immune sentinels (e.g. microglia, macrophages, dendritic cells) that routinely serve as first responders to these infections. The mechanisms that underlie the dynamic programming of these cells following CNS viral infection remain undefined. To gain insights into this programming, we utilized a combination of genomic and two-photon imaging approaches to study a pure innate immune response to a noncytopathic virus (lymphocytic choriomeningitis virus) as it established persistence in the brain. This enabled us to evaluate how global gene expression patterns were translated into myeloid cell dynamics following infection. Two-photon imaging studies revealed that innate myeloid cells mounted a vigorous early response to viral infection characterized by enhanced vascular patrolling and a complete morphological transformation. Interestingly, innate immune activity subsided over time and returned to a quasi-normal state as the virus established widespread persistence in the brain. At the genomic level, early myeloid cell dynamics were associated with massive changes in CNS gene expression, most of which declined over time and were linked to type I interferon signaling (IFN-I). Surprisingly, in the absence of IFN-I signaling, almost no differential gene expression was observed in the nervous system despite increased viral loads. In addition, two-photon imaging studies revealed that IFN-I receptor deficient myeloid cells were unresponsive to viral infection and remained in a naïve state. These data demonstrate that IFN-I engages non-redundant programming responsible for nearly all innate immune activity in the brain following a noncytopathic viral infection. This Achilles' heel could explain why so many neurotropic viruses have acquired strategies to suppress IFN-I.


Assuntos
Regulação da Expressão Gênica , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Células Mieloides/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Animais , Interferon Tipo I/genética , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Knockout , Células Mieloides/patologia , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/genética
18.
PLoS Pathog ; 9(5): e1003362, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696736

RESUMO

Foxp3+ regulatory T (Treg) cells are essential for the maintenance of immune homeostasis and tolerance. During viral infections, Treg cells can limit the immunopathology resulting from excessive inflammation, yet potentially inhibit effective antiviral T cell responses and promote virus persistence. We report here that the fast-replicating LCMV strain Docile triggers a massive expansion of the Treg population that directly correlates with the size of the virus inoculum and its tendency to establish a chronic, persistent infection. This Treg cell proliferation was greatly enhanced in IL-21R-/- mice and depletion of Treg cells partially rescued defective CD8+ T cell cytokine responses and improved viral clearance in some but not all organs. Notably, IL-21 inhibited Treg cell expansion in a cell intrinsic manner. Moreover, experimental augmentation of Treg cells driven by injection of IL-2/anti-IL-2 immune complexes drastically impaired the functionality of the antiviral T cell response and impeded virus clearance. As a consequence, mice became highly susceptible to chronic infection following exposure to low virus doses. These findings reveal virus-driven Treg cell proliferation as potential evasion strategy that facilitates T cell exhaustion and virus persistence. Furthermore, they suggest that besides its primary function as a direct survival signal for antiviral CD8+ T cells during chronic infections, IL-21 may also indirectly promote CD8+ T cell poly-functionality by restricting the suppressive activity of infection-induced Treg cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Interleucinas/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Doença Crônica , Interleucinas/genética , Interleucinas/metabolismo , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Camundongos Knockout , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/virologia
19.
J Immunol ; 191(1): 228-37, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23709682

RESUMO

Clinical evidence for a more active immune response in humans compared with our closest hominid relative, the chimpanzee, includes the progression of HIV infection to AIDS, hepatitis B- and C-related inflammation, autoimmunity, and unwanted harmful immune responses to viral gene transfer vectors. Humans have a unique mutation of the enzyme CMP-N-acetylneuraminic acid hydroxylase (CMAH), causing loss of expression of the sialic acid Neu5Gc. This mutation, occurring 2 million years ago, likely altered the expression and function of ITIM-bearing inhibitory receptors (Siglecs) that bind sialic acids. Previous work showed that human T cells proliferate faster than chimpanzee T cells upon equivalent stimulation. In this article, we report that Cmah(-/-) mouse T cells proliferate faster and have greater expression of activation markers than wild-type mouse T cells. Metabolically reintroducing Neu5Gc diminishes the proliferation and activation of both human and murine Cmah(-/-) T cells. Importantly, Cmah(-/-) mice mount greater T cell responses to an adenovirus encoding an adeno-associated virus capsid transgene. Upon lymphocytic choriomeningitis virus infection, Cmah(-/-) mice make more lymphocytic choriomeningitis virus-specific T cells than WT mice, and these T cells are more polyfunctional. Therefore, a uniquely human glycosylation mutation, modeled in mice, leads to a more proliferative and active T cell population. These findings in a human-like mouse model have implications for understanding the hyperimmune responses that characterize some human diseases.


Assuntos
Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Proliferação de Células , Células Cultivadas , Dependovirus/genética , Dependovirus/imunologia , Dependovirus/metabolismo , Glicosilação , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Subpopulações de Linfócitos T/enzimologia , Regulação para Cima/genética , Regulação para Cima/imunologia
20.
Acta Virol ; 57(2): 160-70, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23600874

RESUMO

Lymphocytic choriomeningitis virus (LCMV) attracts significant attention both as an important experimental model system to study acute and persistent viral infections, and as a neglected human pathogen of clinical significance. This review focuses on the basic aspects and recent advances in the molecular and cell biology of LCMV, the outcome of LCMV infection on its natural host with an emphasis on persistent infection and the outcome of LCMV infection in humans. Lastly, we summarize our contribution to current knowledge on LCM virus.


Assuntos
Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Animais , Humanos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA