Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
PLoS One ; 8(1): e53273, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308183

RESUMO

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.


Assuntos
Glicoproteínas/metabolismo , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/crescimento & desenvolvimento , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Glicoproteínas/genética , Glicosilação , Humanos , Coriomeningite Linfocítica/veterinária , Vírus da Coriomeningite Linfocítica/genética , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Mutação , Neurônios/virologia , Polissacarídeos/genética , Polissacarídeos/metabolismo , Tropismo , Proteínas Virais/genética
11.
Virology ; 436(1): 127-33, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23218200

RESUMO

Maturation of the arenavirus GP precursor (GPC) involves proteolytic processing by cellular signal peptidase and the proprotein convertase subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P), yielding a tripartite complex comprised of a stable signal peptide (SSP), the receptor-binding GP1, and the fusion-active transmembrane GP2. Here we investigated the roles of SKI-1/S1P processing and SSP in the biosynthesis of the recombinant GP ectodomains of lymphocytic choriomeningitis virus (LCMV) and Lassa virus (LASV). When expressed in mammalian cells, the LCMV and LASV GP ectodomains underwent processing by SKI-1/S1P, followed by dissociation of GP1 from GP2. The GP2 ectodomain spontaneously formed trimers as revealed by chemical cross-linking. The endogenous SSP, known to be crucial for maturation and transport of full-length arenavirus GPC was dispensable for processing and secretion of the soluble GP ectodomain, suggesting a specific role of SSP in the stable prefusion conformation and transport of full-length GPC.


Assuntos
Glicoproteínas/química , Glicoproteínas/metabolismo , Vírus Lassa/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Pró-Proteína Convertases/metabolismo , Sinais Direcionadores de Proteínas , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Glicoproteínas/biossíntese , Células HEK293 , Humanos , Proteólise , Proteínas do Envelope Viral/biossíntese
12.
J Virol ; 86(19): 10866-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22787223

RESUMO

The transfer of T cell receptor (TCR) genes by viral vectors represents a promising technique to generate antigen-specific T cells for adoptive immunotherapy. TCR-transduced T cells specific for infectious pathogens have been described, but their protective function in vivo has not yet been examined. Here, we demonstrate that CD8 T cells transduced with the P14 TCR specific for the gp33 epitope of lymphocytic choriomeningitis virus exhibit protective activities in both viral and bacterial infection models in mice.


Assuntos
Antígenos Virais/química , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Glicoproteínas/química , Vírus da Coriomeningite Linfocítica/metabolismo , Fragmentos de Peptídeos/química , Receptores de Antígenos de Linfócitos T/genética , Proteínas Virais/química , Animais , Antígenos/metabolismo , Membrana Celular/metabolismo , Citocinas/metabolismo , Citometria de Fluxo/métodos , Interferon gama/metabolismo , Listeria monocytogenes/metabolismo , Linfócitos/citologia , Camundongos , Fenótipo , Receptores de Antígenos de Linfócitos T/metabolismo , Retroviridae/metabolismo , Baço/metabolismo
13.
J Virol ; 86(18): 9782-93, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761378

RESUMO

The induction of strong CD8(+) T-cell responses against infectious diseases and cancer has remained a major challenge. Depending on the source of antigen and the infectious agent, priming of CD8(+) T cells requires direct and/or cross-presentation of antigenic peptides on major histocompatibility complex (MHC) class I molecules by professional antigen-presenting cells (APCs). However, both pathways show distinct preferences concerning antigen stability. Whereas direct presentation was shown to efficiently present peptides derived from rapidly degraded proteins, cross-presentation is dependent on long-lived antigen species. In this report, we analyzed the role of antigen stability on DNA vaccination and recombinant vaccinia virus (VV) infection using altered versions of the same antigen. The long-lived nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) can be targeted for degradation by N-terminal fusion to ubiquitin or, as we show here, to the ubiquitin-like modifier FAT10. Direct presentation by cells either transfected with NP-encoding plasmids or infected with recombinant VV in vitro was enhanced in the presence of short-lived antigens. In vivo, however, the highest induction of NP-specific CD8(+) T-cell responses was achieved in the presence of long-lived NP. Our experiments provide evidence that targeting antigens for proteasomal degradation does not improve the immunogenicity of DNA vaccines and recombinant VVs. Rather, it is the long-lived antigen that is superior for the efficient activation of MHC class I-restricted immune responses in vivo. Hence, our results suggest a dominant role for antigen cross-priming in DNA vaccination and recombinant VV infection.


Assuntos
Antígenos Virais/metabolismo , Linfócitos T CD8-Positivos/imunologia , Vacinas de DNA/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Animais , Apresentação de Antígeno , Antígenos Virais/genética , Sequência de Bases , Linhagem Celular , Reações Cruzadas , DNA Viral/genética , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/metabolismo , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nucleoproteínas/genética , Nucleoproteínas/imunologia , Nucleoproteínas/metabolismo , Estabilidade Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina/genética , Ubiquitina/imunologia , Ubiquitina/metabolismo , Ubiquitinas/genética , Ubiquitinas/imunologia , Ubiquitinas/metabolismo , Vacinas de DNA/genética , Vaccinia virus/patogenicidade , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
14.
J Virol ; 86(15): 7728-38, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22532683

RESUMO

Arenaviruses perturb innate antiviral defense by blocking induction of type I interferon (IFN) production. Accordingly, the arenavirus nucleoprotein (NP) was shown to block activation and nuclear translocation of interferon regulatory factor 3 (IRF3) in response to virus infection. Here, we sought to identify cellular factors involved in innate antiviral signaling targeted by arenavirus NP. Consistent with previous studies, infection with the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) prevented phosphorylation of IRF3 in response to infection with Sendai virus, a strong inducer of the retinoic acid-inducible gene I (RIG-I)/mitochondrial antiviral signaling (MAVS) pathway of innate antiviral signaling. Using a combination of coimmunoprecipitation and confocal microscopy, we found that LCMV NP associates with the IκB kinase (IKK)-related kinase IKKε but that, rather unexpectedly, LCMV NP did not bind to the closely related TANK-binding kinase 1 (TBK-1). The NP-IKKε interaction was highly conserved among arenaviruses from different clades. In LCMV-infected cells, IKKε colocalized with NP but not with MAVS located on the outer membrane of mitochondria. LCMV NP bound the kinase domain (KD) of IKKε (IKBKE) and blocked its autocatalytic activity and its ability to phosphorylate IRF3, without undergoing phosphorylation. Together, our data identify IKKε as a novel target of arenavirus NP. Engagement of NP seems to sequester IKKε in an inactive complex. Considering the important functions of IKKε in innate antiviral immunity and other cellular processes, the NP-IKKε interaction likely plays a crucial role in arenavirus-host interaction.


Assuntos
Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Linhagem Celular Tumoral , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , RNA Helicases DEAD-box/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Imunidade Inata/genética , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/imunologia , Membranas Mitocondriais/imunologia , Membranas Mitocondriais/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/imunologia , Fosforilação/genética , Fosforilação/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Imunológicos , Vírus Sendai/genética , Vírus Sendai/imunologia , Vírus Sendai/metabolismo
15.
J Virol ; 86(8): 4578-85, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22345463

RESUMO

Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose a significant public health concern in regions where they are endemic. On the other hand, evidence indicates that the globally distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway participates in many cellular processes, including cell survival and differentiation, and also has been shown to play important roles in different steps of the life cycles of a variety of viruses. Here we report that the inhibition of the PI3K/Akt pathway inhibited budding and to a lesser extent RNA synthesis, but not cell entry, of LCMV. Accordingly, BEZ-235, a PI3K inhibitor currently in cancer clinical trials, inhibited LCMV multiplication in cultured cells. These findings, together with those previously reported for Junin virus (JUNV), indicate that targeting the PI3K/Akt pathway could represent a novel antiviral strategy to combat human-pathogenic arenaviruses.


Assuntos
Arenavirus/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Liberação de Vírus , Animais , Arenavirus/efeitos dos fármacos , Arenavirus/genética , Linhagem Celular , Chlorocebus aethiops , Cromonas/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Vírus Lassa/efeitos dos fármacos , Vírus Lassa/genética , Vírus Lassa/metabolismo , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/genética , Vírus da Coriomeningite Linfocítica/metabolismo , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Quinolinas/farmacologia , RNA Viral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
16.
J Immunol ; 187(4): 1634-42, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21742975

RESUMO

Previous studies have identified the inhibitory role that the programmed death 1 (PD-1) pathway plays during chronic infection. Blockade of this pathway results in rescue of viral-specific CD8 T cells, as well as reduction of viral loads in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). We tested the effect of combining PD ligand 1 (PD-L1) blockade with an agonistic regimen that induces 4-1BB costimulation during chronic LCMV infection. There is a boosting effect in the rescue of LCMV-specific CD8 T cell responses after dual treatment with PD-L1 blockade and 4-1BB agonistic Abs when the amount and timing of 4-1BB costimulation are carefully controlled. When PD-L1-blocking Abs are given together with a single low dose of anti-4-1BB agonistic Abs, there is an enhanced and stable expansion of viral-specific CD8 T cells. Conversely, when blocking Abs to PD-L1 are given with a repetitive high dose of anti-4-1BB, there is an initial synergistic expansion of viral-specific CD8 T cells by day 7, followed by dramatic apoptosis by day 14. Viral control paralleled CD8 T cell kinetics after dual treatment. By day 7 posttreatment, viral titers were lower in both of the combined regimens (compared with PD-L1 blockade alone). However, whereas the high dose of anti-4-1BB plus PD-L1 blockade resulted in rebound of viral titers to original levels, the low dose of anti-4-1BB plus PD-L1 blockade resulted in a stable reduction of viral loads. These findings demonstrate the importance of carefully manipulating the balance between activating and inhibitory signals to enhance T cell responses during chronic infection.


Assuntos
Antígeno B7-1/imunologia , Linfócitos T CD8-Positivos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Glicoproteínas de Membrana/imunologia , Peptídeos/imunologia , Transdução de Sinais/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Linfócitos T CD8-Positivos/metabolismo , Doença Crônica , Relação Dose-Resposta a Droga , Feminino , Coriomeningite Linfocítica/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Camundongos , Peptídeos/antagonistas & inibidores , Peptídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Carga Viral
17.
Proc Natl Acad Sci U S A ; 108(7): 2969-74, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21270335

RESUMO

Arenaviruses are a major cause of hemorrhagic fevers endemic to Sub-Saharan Africa and South America, and thus a major public health and medical concern. The prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is widely used as a model system for studying persistent and acute infections, as well as for gaining an understanding of mammalian immune function. When originally characterized three decades ago, the LCMV isolate, Armstrong, which causes an acute infection in adult mice, was found to differ from the LCMV Clone 13 strain that causes a persistent infection by two amino acid changes, one within the virus surface glycoprotein (GP1: F260L) and the other within the virus L polymerase (K1076Q). Mutation F260L was considered solely responsible for the exceptionally strong binding affinity of Clone 13 (L at GP1 260) to its cellular receptor, α-dystroglycan, which among cells of the immune system is preferentially expressed on dendritic cells, and consequently, alters dendritic cell function leading to viral persistence. Recently, we noted a previously overlooked nucleotide difference between these two strains that results in an additional amino acid change in GP1, N176D. To investigate the potential contribution of this newly identified mutation to the Clone 13 phenotype, we used reverse-genetics approaches to generate recombinant LCM viruses with each of these individual mutations. Phenotypic characterization of these rLCMV showed that mutation F260L, but not N176D, in the GP1 of LCMV is essential for mediating the long-term persistence of Clone 13 infections. This work emphasizes the importance of subtle differences in viral strains that determine disease outcomes.


Assuntos
Células Dendríticas/virologia , Vírus da Coriomeningite Linfocítica/genética , Fenótipo , Mutação Puntual/genética , Proteínas Virais de Fusão/genética , Animais , Linhagem Celular , Células Dendríticas/metabolismo , Distroglicanas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Medições Luminescentes , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Internalização do Vírus
18.
J Virol ; 85(4): 1662-70, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21106748

RESUMO

Arenaviruses are enveloped RNA viruses with a nonlytic life cycle that cause acute and persistent infections. Here, we investigated the role of the host cell's unfolded protein response (UPR) in infection of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). In mammalian cells, the endoplasmic reticulum (ER) chaperone protein GRP78/BiP functions as the principal sensor for the induction of the UPR and interacts with three mediators: kinase/endonuclease inositol-requiring protein 1 (IRE1), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Acute infection with LCMV resulted in a selective induction of the ATF6-regulated branch of the UPR, whereas pathways controlled by PERK and IRE1 were neither activated nor blocked. Expression of individual LCMV proteins revealed that the viral glycoprotein precursor (GPC), but not that of other viral proteins, was responsible for the induction of ATF6. Rapid downregulation of the viral GPC during transition from acute to persistent LCMV infection restored basal levels of UPR signaling. To address a possible role of ATF6 signaling in LCMV infection, we used cells deficient in site 2 protease (S2P), a metalloprotease required for the activation of ATF6. Cells deficient in S2P showed significantly lower levels of production of infectious virus during acute but not persistent infection, indicating a requirement for ATF6-mediated signaling for optimal virus multiplication. In summary, acute LCMV infection seems to selectively induce the ATF6-regulated branch of the UPR that is likely beneficial for virus replication and cell viability, but it avoids induction of PERK and IRE1, whose activation may be detrimental for virus and the host cell.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Células Epiteliais/virologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Precursores de Proteínas/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas do Envelope Viral/farmacologia , Fator 6 Ativador da Transcrição/farmacologia , Animais , Arenavirus/patogenicidade , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Proteínas de Choque Térmico/metabolismo , Humanos , Fígado/citologia , Fígado/virologia , Pulmão/citologia , Pulmão/virologia , Vírus da Coriomeningite Linfocítica/metabolismo , Dobramento de Proteína , Precursores de Proteínas/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas/fisiologia , Proteínas do Envelope Viral/metabolismo
19.
J Biol Chem ; 285(24): 18918-27, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20395300

RESUMO

Granule-associated perforin and granzymes (gzms) are key effector molecules of cytotoxic T lymphocytes (Tc cells) and natural killer cells and play a critical role in the control of intracellular pathogens and cancer. Based on the notion that many gzms, including A, B, C, K, H, and M exhibit cytotoxic activity in vitro, all gzms are believed to serve a similar function in vivo. However, more recent evidence supports the concept that gzms are not unidimensional but, rather, possess non-cytotoxic potential, including stimulation of pro-inflammatory cytokines and anti-viral activities. The present study shows that isolated mouse gzmB cleaves the actin-severing mouse protein, cytoplasmic gelsolin (c-gelsolin) in vitro. However, when delivered to intact target cells by ex vivo immune Tc cells, gzmB mediates c-gelsolin proteolysis via activation of caspases 3/7. The NH(2)-terminal c-gelsolin fragment generated by either gzmB or caspase 3 in vitro constitutively severs actin filaments without destroying the target cells. The observation that gzmB secreted by Tc cells initiates a caspase cascade that disintegrates the actin cytoskeleton in target cells suggests that this intracellular process may contribute to anti-viral host defense.


Assuntos
Caspase 3/metabolismo , Citoesqueleto/metabolismo , Gelsolina/química , Granzimas/metabolismo , Linfócitos T Citotóxicos/metabolismo , Animais , Apoptose , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Fibroblastos/metabolismo , Gelsolina/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Camundongos , Microscopia de Fluorescência/métodos , Modelos Biológicos , RNA Mensageiro/metabolismo , Transcrição Gênica
20.
Biochem Biophys Res Commun ; 393(4): 625-30, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20152808

RESUMO

The promyelocytic leukemia protein (PML) forms nuclear bodies (NB) that can be redistributed by virus infection. In particular, lymphocytic choriomeningitis virus (LCMV) influences disruption of PML NB through the interaction of PML with the arenaviral Z protein. In a previous report, we have shown that the disulfide compound NSC20625 has antiviral and virucidal properties against arenaviruses, inducing unfolding and oligomerization of Z without affecting cellular RING-containing proteins such as the PML. Here, we further studied the effect of the zinc-finger-reactive disulfide NSC20625 on PML-Z interaction. In HepG2 cells infected with LCMV or transiently transfected with Z protein constructs, treatment with NSC20625 restored PML distribution from a diffuse-cytoplasmic pattern to punctate, discrete NB which appeared identical to NB found in control, uninfected cells. Similar results were obtained in cells transfected with a construct expressing a Z mutant in zinc-binding site 2 of the RING domain, confirming that this Z-PML interaction requires the integrity of only one zinc-binding site. Altogether, these results show that the compound NSC20625 suppressed Z-mediated PML NB disruption and may be used as a tool for designing novel antiviral strategies against arenavirus infection.


Assuntos
Antivirais/farmacologia , Infecções por Arenaviridae/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Dissulfetos/farmacologia , Guanidinas/farmacologia , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Proteínas Supressoras de Tumor/antagonistas & inibidores , Infecções por Arenaviridae/virologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Vírus da Coriomeningite Linfocítica/metabolismo , Proteínas Nucleares/metabolismo , Proteína da Leucemia Promielocítica , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA