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.
Artículo en Inglés | MEDLINE | ID: mdl-37172825

RESUMEN

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.


Asunto(s)
Leucemia Eritroblástica Aguda , Leucemia Experimental , Ratones , Animales , Virus de la Leucemia Murina de Friend/fisiología , Linfocitos T CD8-positivos , Transducción de Señal , Leucemia Experimental/patología
2.
Front Immunol ; 12: 809774, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35126368

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Interferón-alfa/farmacología , Infecciones por Retroviridae/tratamiento farmacológico , Infecciones por Retroviridae/virología , Animales , Biomarcadores , Línea Celular , Citotoxicidad Inmunológica/efectos de los fármacos , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Femenino , Virus de la Leucemia Murina de Friend/fisiología , Interacciones Huésped-Patógeno/inmunología , Factores Inmunológicos/farmacología , Interferón Tipo I/farmacología , Interferón beta/farmacología , Virus de la Leucemia Murina/fisiología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones , Infecciones por Retroviridae/inmunología , Resultado del Tratamiento , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
3.
PLoS Pathog ; 16(3): e1008340, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32226027

RESUMEN

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.


Asunto(s)
Anticuerpos/administración & dosificación , Melanoma/inmunología , Melanoma/terapia , Infecciones por Retroviridae/inmunología , Linfocitos T Reguladores/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Virus de la Leucemia Murina de Friend/fisiología , Humanos , Inmunoterapia/efectos adversos , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Retroviridae/patología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
4.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434732

RESUMEN

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.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Glicoproteínas de Membrana/inmunología , Infecciones por Retroviridae/inmunología , Receptor Toll-Like 7/inmunología , Infecciones Tumorales por Virus/inmunología , Traslado Adoptivo , Animales , Subgrupos de Linfocitos B/virología , Inmunidad Innata , Interferón Tipo I/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Infecciones por Retroviridae/virología , Transducción de Señal/inmunología , Receptor Toll-Like 7/genética , Infecciones Tumorales por Virus/virología , Replicación Viral
5.
Retrovirology ; 15(1): 68, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30292240

RESUMEN

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.


Asunto(s)
Virus de la Leucemia Murina de Friend/fisiología , Memoria Inmunológica , Células Asesinas Naturales/inmunología , Infecciones por Retroviridae/inmunología , Traslado Adoptivo , Animales , Antígenos Virales/inmunología , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Células Asesinas Naturales/citología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Ratones , Ratones Endogámicos C57BL , Fenotipo
6.
PLoS One ; 13(2): e0192541, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29425215

RESUMEN

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.


Asunto(s)
Células Dendríticas/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Inmunidad Celular/fisiología , Células Th2/inmunología , Animales , Citocinas/metabolismo , Células Dendríticas/metabolismo , Inmunofenotipificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteoma
7.
Sci Rep ; 7(1): 17236, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29222473

RESUMEN

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.


Asunto(s)
Eritroblastos/citología , Virus de la Leucemia Murina de Friend/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/inmunología , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo , Animales , Recuento de Células , Proliferación Celular , Activación Enzimática , Técnicas de Inactivación de Genes , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Bazo/inmunología
8.
J Virol ; 91(23)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28904197

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/antagonistas & inhibidores , Receptores Coestimuladores e Inhibidores de Linfocitos T/genética , Expresión Génica , Infecciones por VIH/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/efectos de los fármacos , Enterotoxinas/farmacología , Virus de la Leucemia Murina de Friend/fisiología , Infecciones por VIH/virología , Humanos , Interferón gamma/biosíntesis , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Infecciones por Retroviridae/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Retrovirology ; 14(1): 25, 2017 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-28415995

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Citidina Desaminasa/metabolismo , Virus de la Leucemia Murina de Friend/inmunología , Interferón Tipo I/metabolismo , Transducción de Señal , Replicación Viral , Animales , Virus de la Leucemia Murina de Friend/fisiología , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia
10.
PLoS One ; 11(11): e0166322, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27880772

RESUMEN

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.


Asunto(s)
Virus de la Leucemia Murina de Friend/fisiología , Interferón gamma/metabolismo , Interleucina-12/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 7/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/virología , Células de la Médula Ósea/citología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Imidazoles/farmacología , Inmunoglobulina G/metabolismo , Interferón gamma/análisis , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/genética
11.
Virology ; 499: 136-143, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27657834

RESUMEN

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.


Asunto(s)
Retrovirus Endógenos/fisiología , Virus de la Leucemia Murina de Friend/fisiología , Interacciones Microbianas , Activación Viral , Animales , Línea Celular , Ratones , Provirus/fisiología , Recombinación Genética , Infecciones por Retroviridae/virología , Análisis de Secuencia de ADN , Carga Viral , Tropismo Viral
12.
Retrovirology ; 13(1): 45, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27363431

RESUMEN

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.


Asunto(s)
Desaminasa APOBEC-3G/metabolismo , Infecciones por VIH/virología , VIH-1/genética , Polimorfismo Genético , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/genética , Desaminasa APOBEC-3G/genética , Alelos , Animales , Modelos Animales de Enfermedad , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/fisiología , Humanos , Ratones , Ratones Transgénicos , Mutación , Replicación Viral
13.
Sci Rep ; 6: 20425, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26846717

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Glicoproteínas de Membrana/metabolismo , Infecciones por Retroviridae/patología , Enfermedad Aguda , Animales , Antígenos CD/genética , Antígeno B7-1/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Células Dendríticas/citología , Células Dendríticas/metabolismo , Virus de la Leucemia Murina de Friend/genética , Interleucina-15/metabolismo , Interleucina-2/análisis , Interleucina-2/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Células 3T3 NIH , Fenotipo , ARN Viral/sangre , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/veterinaria , Replicación Viral
14.
Sci Rep ; 5: 10501, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25994622

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Leucemia Murina de Friend/patogenicidad , Interleucina-6/metabolismo , Infecciones por Retroviridae/patología , Receptores Toll-Like/agonistas , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Virus de la Leucemia Murina de Friend/fisiología , Inmunidad Celular , Interferón gamma/farmacología , Interleucina-6/genética , Interleucina-6/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Replicación Viral
15.
Proc Biol Sci ; 282(1798): 20141568, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25392466

RESUMEN

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.


Asunto(s)
Virus de la Leucemia Murina de Friend/fisiología , Virus de la Leucemia Murina de Friend/patogenicidad , Aptitud Genética , Genotipo , Interacciones Huésped-Patógeno/genética , Leucemia Experimental/genética , Infecciones por Retroviridae/genética , Infecciones Tumorales por Virus/genética , Animales , Evolución Biológica , Femenino , Leucemia Experimental/virología , Ratones , Ratones Endogámicos , Infecciones por Retroviridae/virología , Organismos Libres de Patógenos Específicos , Infecciones Tumorales por Virus/virología , Virulencia/fisiología
16.
Virology ; 468-470: 601-608, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25303118

RESUMEN

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.


Asunto(s)
Citidina Desaminasa/metabolismo , Virus de la Leucemia Murina de Friend/fisiología , Leucemia Experimental/virología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Desaminasas APOBEC-1 , Animales , Citidina Desaminasa/genética , Virus de la Leucemia Murina de Friend/genética , Regulación de la Expresión Génica/fisiología , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/metabolismo , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/metabolismo , Carga Viral , Viremia , Replicación Viral
17.
Virology ; 468-470: 532-544, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25262471

RESUMEN

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.


Asunto(s)
Colobus , Citidina Desaminasa/metabolismo , VIH-1/genética , VIH-1/fisiología , Replicación Viral/fisiología , Animales , Línea Celular , Citidina Desaminasa/genética , Virus de la Leucemia Murina de Friend/fisiología , Regulación Enzimológica de la Expresión Génica , Humanos , Mutación , Estructura Terciaria de Proteína , Proteínas Recombinantes
19.
J Immunol ; 191(12): 6178-90, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24227786

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Endoteliales/efectos de los fármacos , Interleucina-12/metabolismo , Lipopéptidos/farmacología , Hígado/citología , Receptor Toll-Like 2/agonistas , Receptores Toll-Like/agonistas , Traslado Adoptivo , Animales , Apoptosis , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/genética , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica , Inmunidad Celular , Técnicas In Vitro , Interleucina-12/biosíntesis , Interleucina-12/genética , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vacunas de ADN , Replicación Viral
20.
Cell Rep ; 4(4): 689-96, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23972988

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP Monoméricas/metabolismo , Replicación Viral , Animales , Células Dendríticas/metabolismo , Células Dendríticas/virología , Desoxirribonucleótidos/metabolismo , Virus de la Leucemia Murina de Friend/metabolismo , Virus de la Leucemia Murina de Friend/fisiología , VIH-1/metabolismo , VIH-1/fisiología , Interferón beta/genética , Interferón beta/metabolismo , Linfocitos/metabolismo , Linfocitos/virología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transcripción Reversa , Proteína 1 que Contiene Dominios SAM y HD , Transcripción Genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA