Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
1.
Virol Sin ; 38(3): 429-439, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37172825

RESUMO

Erythroleukemia belongs to acute myeloid leukemia (AML) type 6 (M6), and treatment remains difficult due to the poor prognosis of the disease. Friend virus (FV) is a complex of two viruses: Friend murine leukemia virus (F-MuLV) strain along with a defective spleen focus-forming virus (SFFV), which can induce acute erythroleukemia in mice. We have previously reported that activation of vagal α7 nicotinic acetylcholine receptor (nAChR) signaling promotes HIV-1 transcription. Whether vagal muscarinic signaling mediates FV-induced erythroleukemia and the underlying mechanisms remain unclear. In this study, sham and vagotomized mice were intraperitoneally injected with FV. FV infection caused anemia in sham mice, and vagotomy reversed this change. FV infection increased erythroblasts ProE, EryA, and EryB cells in the spleen, and these changes were blocked by vagotomy. In bone marrow, FV infection reduced EryC cells in sham mice, an effect that was counteracted by vagotomy. FV infection increased choline acetyltransferase (ChAT) expression in splenic CD4+ and CD8+ T cells, and this change was reversed by vagotomy. Furthermore, the increase of EryA and EryB cells in spleen of FV-infected wild-type mice was reversed after deletion of ChAT in CD4+ T cells. In bone marrow, FV infection reduced EryB and EryC cells in sham mice, whereas lack of ChAT in CD4+ T cells did not affect this change. Activation of muscarinic acetylcholine receptor 4 (mAChR4) by clozapine N-oxide (CNO) significantly increased EryB in the spleen but decreased the EryC cell population in the bone marrow of FV-infected mice. Thus, vagal-mAChR4 signaling in the spleen and bone marrow synergistically promotes the pathogenesis of acute erythroleukemia. We uncover an unrecognized mechanism of neuromodulation in erythroleukemia.


Assuntos
Leucemia Eritroblástica Aguda , Leucemia Experimental , Camundongos , Animais , Vírus da Leucemia Murina de Friend/fisiologia , Linfócitos T CD8-Positivos , Transdução de Sinais , Leucemia Experimental/patologia
2.
Front Immunol ; 12: 809774, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35126368

RESUMO

Type I Interferons (IFNs), including numerous IFNα subtypes and IFNß, are key molecules during innate and adaptive immune responses against viral infections. These cytokines exert various non-redundant biological activities, although binding to the same receptor. Persistent viral infections are often characterized by increased IFN signatures implicating a potential role of type I IFNs in disease pathogenesis. Using the well-established Friend retrovirus (FV) mouse model, we compared the therapeutic efficacy of IFNα11 and IFNß in acute and chronic retroviral infection. We observed a strong antiviral activity of both IFNs during acute FV infection, whereas only IFNα11 and not IFNß could also control persistent FV infection. The therapeutic treatment with IFNα11 induced the expression of antiviral IFN-stimulated genes (ISG) and improved cytotoxic T cell responses. Finally, dysfunctional CD8+ T cells solely regained cytotoxicity after IFNα11 treatment. Our data provide evidence for opposing activities of type I IFNs during chronic retroviral infections. IFNß was shown to be involved in immune dysfunction in chronic infections, whereas IFNα11 had a strong antiviral potential and reactivated exhausted T cells during persistent retroviral infection. In contrast, during acute infection, both type I IFNs were able to efficiently suppress FV replication.


Assuntos
Antivirais/farmacologia , Interferon-alfa/farmacologia , Infecções por Retroviridae/tratamento farmacológico , Infecções por Retroviridae/virologia , Animais , Biomarcadores , Linhagem Celular , Citotoxicidade Imunológica/efeitos dos fármacos , Gerenciamento Clínico , Modelos Animais de Doenças , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Fatores Imunológicos/farmacologia , Interferon Tipo I/farmacologia , Interferon beta/farmacologia , Vírus da Leucemia Murina/fisiologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Infecções por Retroviridae/imunologia , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
PLoS Pathog ; 16(3): e1008340, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32226027

RESUMO

Combination immunotherapy (CIT) is currently applied as a treatment for different cancers and is proposed as a cure strategy for chronic viral infections. Whether such therapies are efficient during an acute infection remains elusive. To address this, inhibitory receptors were blocked and regulatory T cells depleted in acutely Friend retrovirus-infected mice. CIT resulted in a dramatic expansion of cytotoxic CD4+ and CD8+ T cells and a subsequent reduction in viral loads. Despite limited viral replication, mice developed fatal immunopathology after CIT. The pathology was most severe in the gastrointestinal tract and was mediated by granzyme B producing CD4+ and CD8+ T cells. A similar post-CIT pathology during acute Influenza virus infection of mice was observed, which could be prevented by vaccination. Melanoma patients who developed immune-related adverse events under immune checkpoint CIT also presented with expanded granzyme-expressing CD4+ and CD8+ T cell populations. Our data suggest that acute infections may induce immunopathology in patients treated with CIT, and that effective measures for infection prevention should be applied.


Assuntos
Anticorpos/administração & dosagem , Melanoma/imunologia , Melanoma/terapia , Infecções por Retroviridae/imunologia , Linfócitos T Reguladores/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Antígeno B7-H1/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Humanos , Imunoterapia/efeitos adversos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
4.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31434732

RESUMO

Lymph-borne Friend murine leukemia virus (FrMLV) exploits the sentinel macrophages in the draining popliteal lymph node (pLN) to infect highly permissive innate-like B-1 cells and establish infection in mice. The reason for FrMLV sensitivity of B-1 cells and their impact on viral spread is unknown. Here we demonstrate that Toll-like receptor 7 (TLR7) sensing and type I interferon (IFN-I) signaling in B-1 cells contribute to FrMLV susceptibility. FrMLV infection in B-1 cell-deficient mice (bumble; IκBNS dysfunctional) was significantly lower than that in the wild-type mice and was rescued by adoptive transfer of wild-type B-1 cells. This rescue of FrMLV infection in bumble mice was dependent on intact TLR7 sensing and IFN-I signaling within B-1 cells. Analyses of infected cell types revealed that the reduced infection in bumble mice was due predominantly to compromised virus spread to the B-2 cell population. Our data reveal how FrMLV exploits innate immune sensing and activation in the B-1 cell population for infection and subsequent spread to other lymphocytes.IMPORTANCE Viruses establish infection in hosts by targeting highly permissive cell types. The retrovirus Friend murine leukemia virus (FrMLV) infects a subtype of B cells called B-1 cells that permit robust virus replication. The reason for their susceptibility had remained unknown. We found that innate sensing of incoming virus and the ensuing type I interferon response within B-1 cells are responsible for their observed susceptibility. Our data provide insights into how retroviruses coevolved with the host to co-opt innate immune sensing pathways designed to fight virus infections for establishing infection. Understanding early events in viral spread can inform antiviral intervention strategies that prevent the colonization of a host.


Assuntos
Subpopulações de Linfócitos B/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Glicoproteínas de Membrana/imunologia , Infecções por Retroviridae/imunologia , Receptor 7 Toll-Like/imunologia , Infecções Tumorais por Vírus/imunologia , Transferência Adotiva , Animais , Subpopulações de Linfócitos B/virologia , Imunidade Inata , Interferon Tipo I/imunologia , Linfonodos/imunologia , Linfonodos/virologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Infecções por Retroviridae/virologia , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/genética , Infecções Tumorais por Vírus/virologia , Replicação Viral
5.
Retrovirology ; 15(1): 68, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30292240

RESUMO

Traditionally, NK cells belong to the innate immune system and eliminate virus-infected cells through their germline-encoded receptors. However, NK cells were recently reported to possess memory-like functions that were predominantly provided by hepatic NK cells. Memory properties were mainly documented in contact hypersensitivity models or during cytomegalovirus infections. However, the precise role and the physiologic importance of memory-like NK cells during retroviral infections are still under investigation. Here, we show that Friend retrovirus (FV) infection of mice induced a population of phenotypically memory-like NK cells at 28 days post infection. Upon secondary antigen encounter, these NK cells showed an increased production of the pro-inflammatory cytokines IFNγ and TNFα as well as the death ligand FasL in comparison to naïve NK cells. Furthermore, we found an augmented elimination of antigen-matched but not antigen-mismatched target cells by these memory-like NK cells. In adoptive cell transfer experiments, equal antiviral activities of splenic and hepatic memory-like NK cells during the late phase of acute FV infection were found. Our results strongly imply the existence and antiviral activity of spleen and liver memory-like NK cells in FV infection, which efficiently respond upon secondary exposure to retroviral antigens.


Assuntos
Vírus da Leucemia Murina de Friend/fisiologia , Memória Imunológica , Células Matadoras Naturais/imunologia , Infecções por Retroviridae/imunologia , Transferência Adotiva , Animais , Antígenos Virais/imunologia , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Células Matadoras Naturais/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
6.
PLoS One ; 13(2): e0192541, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29425215

RESUMO

The murine Friend virus (FV) retrovirus model has been widely used to study anti-viral immune responses, and virus-induced cancer. Here we analyzed FV immune evasion mechanisms on the level of dendritic cells (DC) essential for the induction of primary adaptive immune responses. Comparative quantitative proteome analysis of FV-infected DC (FV-DC) of different genotypes (BALB/c, C57BL/6) and non-infected DC revealed numerous genotype-independently regulated proteins rergulating metabolic activity, cytoskeletal rearrangements, and antigen processing/presentation. These alterations may promote virion production in FV-DC. Stimulation of FV-DC with LPS resulted in strongly enhanced IL-10 production which was partially responsible for their attenuated T cell (CD4+, CD8+) stimulatory capacity. Stimulated FV-DC induced less IFN-γ production in T cells required for cellular anti-viral responses, but more T helper cell type 2 (Th2)-associated cytokines (IL-4, IL-5, IL-13). We conclude that FV reprograms DC to promote viral spreading and immune deviation by imprinting a largely maturation-resistant, Th2-biased immunophenotype.


Assuntos
Células Dendríticas/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Imunidade Celular/fisiologia , Células Th2/imunologia , Animais , Citocinas/metabolismo , Células Dendríticas/metabolismo , Imunofenotipagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteoma
7.
Sci Rep ; 7(1): 17236, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29222473

RESUMO

Macrophages are the frontline of defence against foreign microorganisms, including bacteria, parasites, and viruses. During acute viral infection, macrophages must invade the inflamed tissue toward low oxygen concentrations, where genetic cellular responses depend on hypoxia-inducible factors (HIF). In the study reported here we investigated the role of HIF-1α in macrophage function during acute retroviral infection. Wild-type and myeloid cell-specific HIF-1α knockout mice were infected with Friend retrovirus (FV), and immune response was analysed 7 and 10 days after infection. FV infection led to increased spleen weight in wild-type and knockout mice, whereas a profound proliferation of erythroblasts was seen only in wild-type mice. The number of spleen-infiltrating macrophages was also significantly lower in knockout animals. Macrophage invasion after FV infection in wild-type mice led to elevated amounts of activated macrophage-stimulating 1 protein that resulted in massive proliferation of erythrocyte precursor cells. This proliferation was absent from knockout mice because of impaired invasion capabilities of HIF-1α-deficient macrophages. Our study elucidated a novel mechanism of FV-induced erythrocyte precursor cell proliferation.


Assuntos
Eritroblastos/citologia , Vírus da Leucemia Murina de Friend/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Animais , Contagem de Células , Proliferação de Células , Ativação Enzimática , Técnicas de Inativação de Genes , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Baço/imunologia
8.
J Virol ; 91(23)2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28904197

RESUMO

Inhibitory receptors have been extensively described for their importance in regulating immune responses in chronic infections and cancers. Blocking the function of inhibitory receptors such as PD-1, CTLA-4, 2B4, Tim-3, and LAG-3 has shown promise for augmenting CD8 T cell activity and boosting pathogen-specific immunity. However, the prevalence of inhibitory receptors on CD4 T cells and their relative influence on CD4 T cell functionality in chronic HIV infection remains poorly described. We therefore determined and compared inhibitory receptor expression patterns of 2B4, CTLA-4, LAG-3, PD-1, and Tim-3 on virus-specific CD4 and CD8 T cells in relation to their functional T cell profile. In chronic HIV infection, inhibitory receptor distribution differed markedly between cytokine-producing T cell subsets with, gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing cells displaying the highest and lowest prevalence of inhibitory receptors, respectively. Blockade of inhibitory receptors differentially affected cytokine production by cells in response to staphylococcal enterotoxin B stimulation. CTLA-4 blockade increased IFN-γ and CD40L production, while PD-1 blockade strongly augmented IFN-γ, interleukin-2 (IL-2), and TNF-α production. In a Friend retrovirus infection model, CTLA-4 blockade in particular was able to improve control of viral replication. Together, these results show that inhibitory receptor distribution on HIV-specific CD4 T cells varies markedly with respect to the functional subset of CD4 T cells being analyzed. Furthermore, the differential effects of receptor blockade suggest novel methods of immune response modulation, which could be important in the context of HIV vaccination or therapeutic strategies.IMPORTANCE Inhibitory receptors are important for limiting damage by the immune system during acute infections. In chronic infections, however, their expression limits immune system responsiveness. Studies have shown that blocking inhibitory receptors augments CD8 T cell functionality in HIV infection, but their influence on CD4 T cells remains unclear. We assessed the expression of inhibitory receptors on HIV-specific CD4 T cells and their relationship with T cell functionality. We uncovered differences in inhibitory receptor expression depending on the CD4 T cell function. We also found differences in functionality of CD4 T cells following blocking of different inhibitory receptors, and we confirmed our results in a Friend virus retroviral model of infection in mice. Our results show that inhibitory receptor expression on CD4 T cells is linked to CD4 T cell functionality and could be sculpted by blockade of specific inhibitory receptors. These data reveal exciting possibilities for the development of novel treatments and immunotherapeutics.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/antagonistas & inibidores , Receptores Coestimuladores e Inibidores de Linfócitos T/genética , Expressão Gênica , Infecções por HIV/imunologia , Animais , Anticorpos/administração & dosagem , Anticorpos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/efeitos dos fármacos , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Enterotoxinas/farmacologia , Vírus da Leucemia Murina de Friend/fisiologia , Infecções por HIV/virologia , Humanos , Interferon gama/biossíntese , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Infecções por Retroviridae/imunologia , Fator de Necrose Tumoral alfa/biossíntese
9.
Retrovirology ; 14(1): 25, 2017 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-28415995

RESUMO

BACKGROUND: APOBEC3/Rfv3 restricts acute Friend retrovirus (FV) infection and promotes virus-specific neutralizing antibody (NAb) responses. Classical Rfv3 studies utilized FV stocks containing lactate-dehydrogenase elevating virus (LDV), a potent type I interferon inducer. Previously, we showed that APOBEC3 is required for the anti-FV activity of exogenous IFN-alpha treatment. Thus, type I interferon receptor (IFNAR) signaling may be required for the APOBEC3/Rfv3 response. RESULTS: To test if the APOBEC3/Rfv3 response is dependent on type I IFN signaling, we infected IFNAR knockout versus IFNAR/APOBEC3 double-knockout mice with FV/LDV or LDV-free FV, and evaluated acute FV infection and subsequent NAb titers. We show that LDV co-infection and type I IFN signaling are not required for innate APOBEC3-mediated restriction. By contrast, removal of LDV and/or type I IFN signaling abrogated the APOBEC3-dependent NAb response. CONCLUSIONS: APOBEC3 can restrict retroviruses in a type I IFN-independent manner in vivo. By contrast, the ability of APOBEC3 to promote NAb responses is type I IFN-dependent. These findings reveal novel insights on the interplay between type I IFNs and APOBEC3 in vivo that may have implications for augmenting antiretroviral NAb responses.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Citidina Desaminase/metabolismo , Vírus da Leucemia Murina de Friend/imunologia , Interferon Tipo I/metabolismo , Transdução de Sinais , Replicação Viral , Animais , Vírus da Leucemia Murina de Friend/fisiologia , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência
10.
PLoS One ; 11(11): e0166322, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27880772

RESUMO

Knowledge of the processes that underlie IgG subclass switching could inform strategies designed to counteract infections and autoimmunity. Here we show that TLR7 ligands induce subsets of memory CD4 and CD8 T cells to secrete interferon γ (IFNγ) in the absence of antigen receptor stimulation. In turn, TLR ligation and IFNγ cause B cells to express the transcription factor, T-bet, and to switch immunoglobulin production to IgG2a/c. Absence of TLR7 in T cells leads to the impaired T-bet expression in B cells and subsequent inefficient IgG2a isotype switching both in vitro and during the infection with Friend virus in vivo. Our results reveal a surprising mechanism of antiviral IgG subclass switching through T-cell intrinsic TLR7/IL-12 signaling.


Assuntos
Vírus da Leucemia Murina de Friend/fisiologia , Interferon gama/metabolismo , Interleucina-12/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 7 Toll-Like/metabolismo , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/virologia , Células da Medula Óssea/citologia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Imidazóis/farmacologia , Imunoglobulina G/metabolismo , Interferon gama/análise , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/genética
11.
Virology ; 499: 136-143, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27657834

RESUMO

We have demonstrated in a mouse model that infection with a retrovirus can lead not only to the generation of recombinants between exogenous and endogenous gammaretrovirus, but also to the mobilization of endogenous proviruses by pseudotyping entire polytropic proviral transcripts and facilitating their infectious spread to new cells. However, the frequency of this occurrence, the kinetics, and the identity of mobilized endogenous proviruses was unclear. Here we find that these mobilized transcripts are detected after only one day of infection. They predominate over recombinant polytropic viruses early in infection, persist throughout the course of disease and are comprised of multiple different polytropic proviruses. Other endogenous retroviral elements such as intracisternal A particles (IAPs) were not detected. The integration of the endogenous transcripts into new cells could result in loss of transcriptional control and elevated expression which may facilitate pathogenesis, perhaps by contributing to the generation of polytropic recombinant viruses.


Assuntos
Retrovirus Endógenos/fisiologia , Vírus da Leucemia Murina de Friend/fisiologia , Interações Microbianas , Ativação Viral , Animais , Linhagem Celular , Camundongos , Provírus/fisiologia , Recombinação Genética , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Carga Viral , Tropismo Viral
12.
Retrovirology ; 13(1): 45, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27363431

RESUMO

Humans encode seven APOBEC3 proteins (A-H), with A3G, 3F and 3H as the major factors restricting HIV-1 replication. HIV-1, however, encodes Vif, which counteracts A3 proteins by chaperoning them to the proteasome where they are degraded. Vif polymorphisms found in HIV-1s isolated from infected patients have varying anti-A3G potency when assayed in vitro, but there are few studies demonstrating this in in vivo models. Here, we created Friend murine leukemia viruses encoding vif alleles that were previously shown to differentially neutralize A3G in cell culture or that were originally found in primary HIV-1 isolates. Infection of transgenic mice expressing different levels of human A3G showed that these naturally occurring Vif variants differed in their ability to counteract A3G during in vivo infection, although the effects on viral replication were not identical to those seen in cultured cells. We also found that the polymorphic Vifs that attenuated viral replication reverted to wild type only in A3G transgenic mice. Finally, we found that the level of A3G-mediated deamination was inversely correlated with the level of viral replication. This animal model should be useful for studying the functional significance of naturally occurring vif polymorphisms, as well as viral evolution in the presence of A3G.


Assuntos
Desaminase APOBEC-3G/metabolismo , Infecções por HIV/virologia , HIV-1/genética , Polimorfismo Genético , Produtos do Gene vif do Vírus da Imunodeficiência Humana/genética , Desaminase APOBEC-3G/genética , Alelos , Animais , Modelos Animais de Doenças , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Replicação Viral
13.
Sci Rep ; 6: 20425, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26846717

RESUMO

Tetherin/BST-2 is a host restriction factor that inhibits retrovirus release from infected cells in vitro by tethering nascent virions to the plasma membrane. However, contradictory data exists on whether Tetherin inhibits acute retrovirus infection in vivo. Previously, we reported that Tetherin-mediated inhibition of Friend retrovirus (FV) replication at 2 weeks post-infection correlated with stronger natural killer, CD4+ T and CD8+ T cell responses. Here, we further investigated the role of Tetherin in counteracting retrovirus replication in vivo. FV infection levels were similar between wild-type (WT) and Tetherin KO mice at 3 to 7 days post-infection despite removal of a potent restriction factor, Apobec3/Rfv3. However, during this phase of acute infection, Tetherin enhanced myeloid dendritic cell (DC) function. DCs from infected, but not uninfected, WT mice expressed significantly higher MHC class II and the co-stimulatory molecule CD80 compared to Tetherin KO DCs. Tetherin-associated DC activation during acute FV infection correlated with stronger NK cell responses. Furthermore, Tetherin+ DCs from FV-infected mice more strongly stimulated FV-specific CD4+ T cells ex vivo compared to Tetherin KO DCs. The results link the antiretroviral and immunomodulatory activity of Tetherin in vivo to improved DC activation and MHC class II antigen presentation.


Assuntos
Antígenos CD/metabolismo , Células Dendríticas/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Glicoproteínas de Membrana/metabolismo , Infecções por Retroviridae/patologia , Doença Aguda , Animais , Antígenos CD/genética , Antígeno B7-1/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Citidina Desaminase/deficiência , Citidina Desaminase/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Vírus da Leucemia Murina de Friend/genética , Interleucina-15/metabolismo , Interleucina-2/análise , Interleucina-2/metabolismo , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Células NIH 3T3 , Fenótipo , RNA Viral/sangue , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/veterinária , Replicação Viral
14.
Sci Rep ; 5: 10501, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-25994622

RESUMO

We have previously shown that Toll-like receptor (TLR) agonists contribute to the control of viral infection by augmenting virus-specific CD8(+) T-cell responses. It is also well established that signaling by TLRs results in the production of pro-inflammatory cytokines such as interleukin 6 (IL-6). However, how these pro-inflammatory cytokines influence the virus-specific CD8(+) T-cell response during the TLR agonist stimulation remained largely unknown. Here, we investigated the role of TLR-induced IL-6 in shaping virus-specific CD8(+) T-cell responses in the Friend retrovirus (FV) mouse model. We show that the TLR agonist induced IL-6 counter-regulates effector CD8(+) T-cell responses. IL-6 potently inhibited activation and cytokine production of CD8(+) T cells in vitro. This effect was mediated by a direct stimulation of CD8(+) T cells by IL-6, which induced upregulation of STAT3 phosphorylation and SOCS3 and downregulated STAT4 phosphorylation and T-bet. Moreover, combining TLR stimulation and IL-6 blockade during an acute FV infection resulted in enhanced virus-specific CD8(+) T-cell immunity and better control of viral replication. These results have implications for our understanding of the role of TLR induced pro-inflammatory cytokines in regulating effector T cell responses and for the development of therapeutic strategies to overcome T cell dysfunction in chronic viral infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Interleucina-6/metabolismo , Infecções por Retroviridae/patologia , Receptores Toll-Like/agonistas , Doença Aguda , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Vírus da Leucemia Murina de Friend/fisiologia , Imunidade Celular , Interferon gama/farmacologia , Interleucina-6/genética , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Replicação Viral
15.
Proc Biol Sci ; 282(1798): 20141568, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25392466

RESUMO

Reduced genetic variation among hosts may favour the emergence of virulent infectious diseases by enhancing pathogen replication and its associated virulence due to adaptation to a limited set of host genotypes. Here, we test this hypothesis using experimental evolution of a mouse-specific retroviral pathogen, Friend virus (FV) complex. We demonstrate rapid fitness (i.e. viral titre) and virulence increases when FV complex serially infects a series of inbred mice representing the same genotype, but not when infecting a diverse array of inbred mouse strains modelling the diversity in natural host populations. Additionally, a single infection of a different host genotype was sufficient to constrain the emergence of a high fitness/high virulence FV complex phenotype in these experiments. The potent inhibition of viral fitness and virulence was associated with an observed loss of the defective retroviral genome (spleen focus-forming virus), whose presence exacerbates infection and drives disease in susceptible mice. Results from our experiments provide an important first step in understanding how genetic variation among vertebrate hosts influences pathogen evolution and suggests that serial exposure to different genotypes within a single host species may act as a constraint on pathogen adaptation that prohibits the emergence of more virulent infections. From a practical perspective, these results have implications for low-diversity host populations such as endangered species and domestic animals.


Assuntos
Vírus da Leucemia Murina de Friend/fisiologia , Vírus da Leucemia Murina de Friend/patogenicidade , Aptidão Genética , Genótipo , Interações Hospedeiro-Patógeno/genética , Leucemia Experimental/genética , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Animais , Evolução Biológica , Feminino , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos , Infecções por Retroviridae/virologia , Organismos Livres de Patógenos Específicos , Infecções Tumorais por Vírus/virologia , Virulência/fisiologia
16.
Virology ; 468-470: 601-608, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25303118

RESUMO

APOBEC1 is a cytidine deaminase involved in cholesterol metabolism that has been linked to retrovirus restriction, analogous to the evolutionarily-related APOBEC3 proteins. In particular, murine APOBEC1 was shown to inhibit Friend retrovirus (FV) in vitro, generating high levels of C-to-T and G-to-A mutations. These observations raised the possibility that FV infection might be altered in APOBEC1-null mice. To examine this question directly, we infected wild-type and APOBEC1-null mice with FV complex and evaluated acute infection levels. Surprisingly, APOBEC1-null mice exhibited similar cellular infection levels and plasma viremia relative to wild-type mice. Moreover, next-generation sequencing analyses revealed that in contrast to APOBEC3, APOBEC1 did not enhance retroviral C-to-T and G-to-A mutational frequencies in genomic DNA. Thus, APOBEC1 neither inhibited nor significantly drove the molecular evolution of FV in vivo. Our findings reinforce that not all retrovirus restriction factors characterized as potent in vitro may be functionally relevant in vivo.


Assuntos
Citidina Desaminase/metabolismo , Vírus da Leucemia Murina de Friend/fisiologia , Leucemia Experimental/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Desaminase APOBEC-1 , Animais , Citidina Desaminase/genética , Vírus da Leucemia Murina de Friend/genética , Regulação da Expressão Gênica/fisiologia , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mutação , Infecções por Retroviridae/genética , Infecções por Retroviridae/metabolismo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/metabolismo , Carga Viral , Viremia , Replicação Viral
17.
Virology ; 468-470: 532-544, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25262471

RESUMO

The APOBEC3 (A3) deaminases are retrovirus restriction factors that were proposed as inhibitory components of HIV-1 gene therapy vectors. However, A3 mutational activity may induce undesired genomic damage and enable HIV-1 to evade drugs and immune responses. Here, we show that A3A protein from Colobus guereza (colA3A) can restrict HIV-1 replication in producer cells in a deaminase-independent manner without inducing DNA damage. Neither HIV-1 reverse transcription nor integration were significantly affected by colA3A, but capsid protein synthesis was inhibited. The determinants for colA3A restriction mapped to the N-terminal region. These properties extend to A3A from mandrills and De Brazza's monkeys. Surprisingly, truncated colA3A proteins expressing only the N-terminal 100 amino acids effectively exclude critical catalytic regions but retained potent cellular restriction activity. These highlight a unique mechanism of cellular HIV-1 restriction by several Old World monkey A3A proteins that may be exploited for functional HIV-1 cure strategies.


Assuntos
Colobus , Citidina Desaminase/metabolismo , HIV-1/genética , HIV-1/fisiologia , Replicação Viral/fisiologia , Animais , Linhagem Celular , Citidina Desaminase/genética , Vírus da Leucemia Murina de Friend/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Mutação , Estrutura Terciária de Proteína , Proteínas Recombinantes
19.
J Immunol ; 191(12): 6178-90, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24227786

RESUMO

Liver sinusoidal endothelial cells (LSECs) are unique organ-resident APCs capable of Ag cross-presentation and subsequent tolerization of naive CD8(+) T cells. Under certain conditions, LSECs can switch from a tolerogenic to an immunogenic state and promote the development of T cell immunity. However, little is known about the mechanisms of LSECs to induce T cell immunity. In this study, we investigated whether functional maturation of LSECs can be achieved by TLR ligand stimulation and elucidated the mechanisms involved in LSEC-induced T cell immunity. We demonstrate that pretreatment of LSECs with palmitoyl-3-cysteine-serine-lysine-4 (P3C; TLR1/2 ligand) but not poly(I:C) (TLR3 ligand) or LPS (TLR4 ligand) reverted their suppressive properties to induce T cell immunity. Importantly, P3C stimulation caused functional maturation of Ag-presenting LSECs and enabled them to activate virus-specific CD8(+) T cells. The LSEC-mediated CD8(+) T cell immunity was initiated by soluble mediators, one of which was IL-12 secreted at a low but sustained level after P3C stimulation. P3C stimulation did not induce programmed death ligand 1 expression on LSECs, thereby favoring T cell proliferation and activation instead of suppression. Our data suggest that LSECs undergo maturation exclusively in response to TLR1/2 ligand stimulation and that the immunological status of LSECs was dependent upon the balance between programmed death ligand 1 and IL-12 expression. These results have implications for our understanding of liver-specific tolerance and autoimmunity and for the development of strategies to overcome T cell tolerance in situations such as chronic viral liver infections or liver cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/efeitos dos fármacos , Interleucina-12/metabolismo , Lipopeptídeos/farmacologia , Fígado/citologia , Receptor 2 Toll-Like/agonistas , Receptores Toll-Like/agonistas , Transferência Adotiva , Animais , Apoptose , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/biossíntese , Antígeno B7-H1/genética , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Tolerância Imunológica , Imunidade Celular , Técnicas In Vitro , Interleucina-12/biossíntese , Interleucina-12/genética , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vacinas de DNA , Replicação Viral
20.
Cell Rep ; 4(4): 689-96, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23972988

RESUMO

Aicardi-Goutières syndrome (AGS), a hereditary autoimmune disease, clinically and biochemically overlaps with systemic lupus erythematosus (SLE) and, like SLE, is characterized by spontaneous type I interferon (IFN) production. The finding that defects of intracellular nucleases cause AGS led to the concept that intracellular accumulation of nucleic acids triggers inappropriate production of type I IFN and autoimmunity. AGS can also be caused by defects of SAMHD1, a 3' exonuclease and deoxynucleotide (dNTP) triphosphohydrolase. Human SAMHD1 is an HIV-1 restriction factor that hydrolyzes dNTPs and decreases their concentration below the levels required for retroviral reverse transcription. We show in gene-targeted mice that also mouse SAMHD1 reduces cellular dNTP concentrations and restricts retroviral replication in lymphocytes, macrophages, and dendritic cells. Importantly, the absence of SAMHD1 triggered IFN-ß-dependent transcriptional upregulation of type I IFN-inducible genes in various cell types indicative of spontaneous IFN production. SAMHD1-deficient mice may be instrumental for elucidating the mechanisms that trigger pathogenic type I IFN responses in AGS and SLE.


Assuntos
Proteínas Monoméricas de Ligação ao GTP/metabolismo , Replicação Viral , Animais , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Desoxirribonucleotídeos/metabolismo , Vírus da Leucemia Murina de Friend/metabolismo , Vírus da Leucemia Murina de Friend/fisiologia , HIV-1/metabolismo , HIV-1/fisiologia , Interferon beta/genética , Interferon beta/metabolismo , Linfócitos/metabolismo , Linfócitos/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Transcrição Reversa , Proteína 1 com Domínio SAM e Domínio HD , Transcrição Gênica , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA