Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 570
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 208(2): 444-453, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34893529

RESUMEN

SAMHD1 is a potent HIV-1 restriction factor that blocks reverse transcription in monocytes, dendritic cells and resting CD4+ T cells by decreasing intracellular dNTP pools. However, SAMHD1 may diminish innate immune sensing and Ag presentation, resulting in a weaker adaptive immune response. To date, the role of SAMHD1 on antiretroviral immunity remains unclear, as mouse SAMHD1 had no impact on murine retrovirus replication in prior in vivo studies. Here, we show that SAMHD1 significantly inhibits acute Friend retrovirus infection in mice. Pretreatment with LPS, a significant driver of inflammation during HIV-1 infection, further unmasked a role for SAMHD1 in influencing immune responses. LPS treatment in vivo doubled the intracellular dNTP levels in immune compartments of SAMHD1 knockout but not wild-type mice. SAMHD1 knockout mice exhibited higher plasma infectious viremia and proviral DNA loads than wild-type mice at 7 d postinfection (dpi), and proviral loads inversely correlated with a stronger CD8+ T cell response. SAMHD1 deficiency was also associated with weaker NK, CD4+ T and CD8+ T cell responses by 14 dpi and weaker neutralizing Ab responses by 28 dpi. Intriguingly, SAMHD1 influenced these cell-mediated immune (14 dpi) and neutralizing Ab (28 dpi) responses in male but not female mice. Our findings formally demonstrate SAMHD1 as an antiretroviral factor in vivo that could promote adaptive immune responses in a sex-dependent manner. The requirement for LPS to unravel the SAMHD1 immunological phenotype suggests that comorbidities associated with a "leaky" gut barrier may influence the antiviral function of SAMHD1 in vivo.


Asunto(s)
Inmunidad Adaptativa/inmunología , Virus de la Leucemia Murina de Friend/crecimiento & desarrollo , Lipopolisacáridos/farmacología , Infecciones por Retroviridae/prevención & control , Proteína 1 que Contiene Dominios SAM y HD/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , ADN Viral/sangre , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Retroviridae/virología , Transcripción Reversa/genética , Proteína 1 que Contiene Dominios SAM y HD/inmunología , Carga Viral
2.
EMBO J ; 37(3): 398-412, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29263148

RESUMEN

To analyze the potential role of Tregs in controlling the TCR repertoire breadth to a non-self-antigen, a TCRß transgenic mouse model (EF4.1) expressing a limited, yet polyclonal naïve T-cell repertoire was used. The response of EF4.1 mice to an I-Ab-associated epitope of the F-MuLV envelope protein is dominated by clones expressing a Vα2 gene segment, thus allowing a comprehensive analysis of the TCRα repertoire in a relatively large cohort of mice. Control and Treg-depleted EF4.1 mice were immunized, and the extent of the Vα2-bearing, antigen-specific TCR repertoire was characterized by high-throughput sequencing and spectratyping analysis. In addition to increased clonal expansion and acquisition of effector functions, Treg depletion led to the expression of a more diverse TCR repertoire comprising several private clonotypes rarely observed in control mice or in the pre-immune repertoire. Injection of anti-CD86 antibodies in vivo led to a strong reduction in TCR diversity, suggesting that Tregs may influence TCR repertoire diversity by modulating costimulatory molecule availability. Collectively, these studies illustrate an additional mechanism whereby Tregs control the immune response to non-self-antigens.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígeno B7-2/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Proteínas del Envoltorio Viral/inmunología
3.
Histochem Cell Biol ; 156(2): 165-182, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34003355

RESUMEN

Diagnosis and prognosis of breast cancer is based on disease staging identified through histopathological and molecular biology techniques. Animal models are used to gain mechanistic insights into the development of breast cancer. C(3)1-TAg is a genetically engineered mouse model that develops mammary cancer. However, carcinogenesis caused by this transgene was characterized in the Friend Virus B (FVB) background. As most genetic studies are done in mice with C57BL/6 J background, we aimed to define the histological alterations in C3(1)-TAg C57BL/6 J animals. Our results showed that C3(1)-TAg animals with C57BL/6 J background develop solid-basaloid adenoid cystic carcinomas with increased fibrosis, decreased area of adipocytes, and a high proliferative index, which are triple-negative for progesterone, estrogen, and human epidermal growth factor receptor 2 (HER2) receptors. Our results also revealed that tumor development is slower in the C57BL/6 J background when compared with the FVB strain, providing a better model to study the different stages in breast cancer progression.


Asunto(s)
Antígenos Virales de Tumores/genética , Neoplasias de la Mama/genética , Carcinoma Adenoide Quístico/genética , Modelos Genéticos , Animales , Antígenos Virales de Tumores/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinoma Adenoide Quístico/inmunología , Carcinoma Adenoide Quístico/patología , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
4.
PLoS Pathog ; 15(9): e1008043, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31568492

RESUMEN

Immunization vectors based on cytomegalovirus (CMV) have attracted a lot of interest in recent years because of their high efficacy in the simian immunodeficiency virus (SIV) macaque model, which has been attributed to their ability to induce strong, unusually broad, and unconventionally restricted CD8+ T cell responses. To evaluate the ability of CMV-based vectors to mediate protection by other immune mechanisms, we evaluated a mouse CMV (MCMV)-based vector encoding Friend virus (FV) envelope (Env), which lacks any known CD8+ T cell epitopes, for its protective efficacy in the FV mouse model. When we immunized highly FV-susceptible mice with the Env-encoding MCMV vector (MCMV.env), we could detect high frequencies of Env-specific CD4+ T cells after a single immunization. While the control of an early FV challenge infection was highly variable, an FV infection applied later after immunization was tightly controlled by almost all immunized mice. Protection of mice correlated with their ability to mount a robust anamnestic neutralizing antibody response upon FV infection, but Env-specific CD4+ T cells also produced appreciable levels of interferon γ. Depletion and transfer experiments underlined the important role of antibodies for control of FV infection but also showed that while no Env-specific CD8+ T cells were induced by the MCMV.env vaccine, the presence of CD8+ T cells at the time of FV challenge was required. The immunity induced by MCMV.env immunization was long-lasting, but was restricted to MCMV naïve animals. Taken together, our results demonstrate a novel mode of action of a CMV-based vaccine for anti-retrovirus immunization that confers strong protection from retrovirus challenge, which is conferred by CD4+ T cells and antibodies.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Muromegalovirus/inmunología , Vacunas Virales/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antivirales/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Femenino , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/patogenicidad , Productos del Gen env/genética , Productos del Gen env/inmunología , Vectores Genéticos , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Muromegalovirus/genética , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
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.
J Virol ; 91(22)2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28904191

RESUMEN

Natural killer (NK) cells are part of the innate immune system and recognize virus-infected cells as well as tumor cells. Conflicting data about the beneficial or even detrimental role of NK cells in different infectious diseases have been described previously. While the type of pathogen strongly influences NK cell functionality, less is known about how the infection dose influences the quality of a NK cell response against retroviruses. In this study, we used the well-established Friend retrovirus (FV) mouse model to investigate the impact of virus dose on the induction of antiviral NK cell functions. High-dose virus inoculation increased initial virus replication compared to that with medium- or low-dose viral challenge and significantly improved NK cell activation. Antiviral NK cell activity, including in vivo cytotoxicity toward infected target cells, was also enhanced by high-dose virus infection. NK cell activation following high-dose viral challenge was likely mediated by activated dendritic cells (DCs) and macrophages and the NK cell-stimulating cytokines interleukin 15 (IL-15) and IL-18. Neutralization of these cytokines decreased NK cell functions and increased viral loads, whereas IL-15 and IL-18 therapy improved NK cell activity. Here we demonstrate that virus dose positively correlates with antiviral NK cell activity and function, which are at least partly driven by IL-15 and IL-18. Our results suggest that NK cell activity may be therapeutically enhanced by administering IL-15 and IL-18 in virus infections that inadequately activate NK cells.IMPORTANCE In infections with retroviruses, like HIV and FV infection of mice, NK cells clearly mediate antiviral activities, but they are usually not sufficient to prevent severe pathology. Here we show that the initial infection dose impacts the induction of an antiviral NK cell response during an acute retroviral infection, which had not investigated before. High-dose infection resulted in a strong NK cell functionality, whereas no antiviral activities were detected after low- or medium-dose infection. Interestingly, DCs and macrophages were highly activated after high-dose FV challenge, which corresponded with increased levels of NK cell-stimulating cytokines IL-15 and IL-18. IL-15 and IL-18 neutralization decreased NK cell functions, whereas IL-15 and IL-18 therapy improved NK cell activity. Here we show the importance of cytokines for NK cell activation in retroviral infections; our findings suggest that immunotherapy combining the well-tolerated cytokines IL-15 and IL-18 might be an interesting approach for antiretroviral treatment.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Infecciones por Retroviridae/inmunología , Animales , Relación Dosis-Respuesta Inmunológica , Femenino , Interleucina-15/inmunología , Interleucina-15/farmacología , Interleucina-18/inmunología , Interleucina-18/farmacología , Ratones , Infecciones por Retroviridae/tratamiento farmacológico
7.
J Virol ; 91(20)2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28768877

RESUMEN

Adenovirus (Ad)-based immunization is a popular approach in vaccine development, and Ad-based vectors are renowned for their potential to induce strong CD8+ T cell responses to the encoded transgene. Surprisingly, we previously found in the mouse Friend retrovirus (FV) model that Ad-based immunization did not induce CD8+ T cell responses to the FV Leader-Gag-derived immunodominant epitope GagL85-93 We show now that induction of GagL85-93-specific CD8+ T cells was highly effective when leader-Gag was delivered by plasmid DNA immunization, implying a role for Ad-derived epitopes in mediating unresponsiveness. By immunizing with DNA constructs encoding strings of GagL85-93 and the two Ad-derived epitopes DNA-binding protein418-426 (DBP418-426) and hexon486-494, we confirmed that Ad epitopes prevent induction of GagL85-93-specific CD8+ T cells. Interestingly, while DBP418-426 did not interfere with GagL85-93-specific CD8+ T cell induction, the H-2Dd-restricted hexon486-494 suppressed the CD8+ T cell response to the H-2Db-restricted GagL85-93 strongly in H-2b/d mice but not in H-2b/b mice. This finding indicates that competition occurs at the level of responding CD8+ T cells, and we could indeed demonstrate that coimmunization with an interleukin 2 (IL-2)-encoding plasmid restored GagL85-93-specific CD8+ T cell responses to epitope strings in the presence of hexon486-494 IL-2 codelivery did not restore GagL85-93 responsiveness in Ad-based immunization, however, likely due to the presence of further epitopes in the Ad vector. Our findings show that seemingly immunodominant transgene epitopes can be dominated by Ad-derived epitopes. These findings underline the importance of thorough characterization of vaccine vectors, and modifications of vectors or immunogens may be required to prevent impaired transgene-specific immune responses.IMPORTANCE Ad-based vectors are widely used in experimental preclinical and clinical immunization studies against numerous infectious agents, such as human immunodeficiency virus, Ebola virus, Plasmodium falciparum, or Mycobacterium tuberculosis Preexisting immunity to Ad-based vectors is widely recognized as a hindrance to the widespread use of Ad-based vectors for immunizations in humans; however, our data show that an immune response to Ad-derived T cell epitopes can also result in loss or impairment of transgene-specific immune responses in prenaive vaccinees due to immune competition. Our results highlight that seemingly immunodominant epitopes may be affected by dominance of vector-derived epitopes, and modifications of the vector design or the immunogens employed in immunization may lead to more effective vaccines.


Asunto(s)
Adenoviridae/genética , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Epítopos Inmunodominantes/inmunología , Transgenes , Adenoviridae/inmunología , Vacunas contra el Adenovirus/inmunología , Animales , Linfocitos T CD8-positivos/química , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/inmunología , Vectores Genéticos , Inmunización , Interleucina-2/administración & dosificación , Interleucina-2/genética , Interleucina-2/inmunología , Activación de Linfocitos , Ratones , Retroviridae/genética , Retroviridae/inmunología , Vacunas de ADN/inmunología
8.
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
9.
Retrovirology ; 14(1): 5, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28122574

RESUMEN

BACKGROUND: Natural killer T cells (NKT cells) play an important role in the immunity against viral infections. They produce cytokines or have direct cytolytic effects that can restrict virus replication. However, the exact function of NKT cells in retroviral immunity is not fully elucidated. Therefore, we analyzed the antiretroviral functions of NKT cells in mice infected with the Friend retrovirus (FV). RESULTS: After FV infection numbers of NKT cells remained unchanged but activation as well as improved effector functions of NKT cells were found. While the release of pro-inflammatory cytokines was not changed after infection, activated NKT cells revealed an elevated cytotoxic potential. Stimulation with α-Galactosylceramide significantly increased not only total NKT cell numbers and activation but also the anti-retroviral capacity of NKT cells. CONCLUSION: We demonstrate a strong activation and a potent cytolytic function of NKT cells during acute retroviral infection. Therapeutic treatment with α-Galactosylceramide could further improve the reduction of early retroviral replication by NKT cells, which could be utilized for future treatment against viral infections.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Células T Asesinas Naturales/inmunología , Infecciones por Retroviridae/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Citocinas/metabolismo , Citotoxicidad Inmunológica , Activación de Linfocitos , Ratones
10.
Retrovirology ; 14(1): 8, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28166802

RESUMEN

BACKGROUND: In the Friend retrovirus mouse model we developed potent adenovirus-based vaccines that were designed to induce either strong Friend virus GagL85-93-specific CD8+ T cell or antibody responses, respectively. To optimize the immunization outcome we evaluated vaccination strategies using combinations of these vaccines. RESULTS: While the vaccines on their own confer strong protection from a subsequent Friend virus challenge, the simple combination of the vaccines for the establishment of an optimized immunization protocol did not result in a further improvement of vaccine effectivity. We demonstrate that the co-immunization with GagL85-93/leader-gag encoding vectors together with envelope-encoding vectors abrogates the induction of GagL85-93-specific CD8+ T cells, and in successive immunization protocols the immunization with the GagL85-93/leader-gag encoding vector had to precede the immunization with an envelope encoding vector for the efficient induction of GagL85-93-specific CD8+ T cells. Importantly, the antibody response to envelope was in fact enhanced when the mice were adenovirus-experienced from a prior immunization, highlighting the expedience of this approach. CONCLUSIONS: To circumvent the immunosuppressive effect of envelope on immune responses to simultaneously or subsequently administered immunogens, we developed a two immunizations-based vaccination protocol that induces strong immune responses and confers robust protection of highly Friend virus-susceptible mice from a lethal Friend virus challenge.


Asunto(s)
Adenoviridae/genética , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Vacunación/métodos , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Adenoviridae/inmunología , Animales , Anticuerpos Antivirales/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/inmunología , Vectores Genéticos , Células HEK293 , Humanos , Ratones , Infecciones por Retroviridae/virología , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/genética , Vacunas Combinadas/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
11.
Retrovirology ; 14(1): 28, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28449719

RESUMEN

BACKGROUND: Retroviral envelope (Env) proteins are known to exhibit immunosuppressive properties, which become apparent not only in retroviral infections, but also in gene-based immunizations using retroviral immunogens, where envelope interferes with the induction of CD8+ T cell responses towards another, simultaneously or subsequently delivered, immunogen. RESULTS: In the Friend retrovirus mouse model, immunization with a plasmid encoding the Friend murine leukemia virus (F-MuLV) Leader-Gag protein resulted in induction of a strong GagL85-93-specific CD8+ T cell response, while the response was completely abrogated by co-immunization with an F-MuLV Env-encoding plasmid. In order to overcome this interference of retroviral envelope, we employed plasmids encoding the cytokines interleukin (IL) 1ß, IL2, IL12, IL15, IL21, IL28A or granulocyte-macrophage colony-stimulating factor (GM-CSF) as genetic adjuvants. Co-application of plasmids encoding IL2, IL12, IL21, IL28A and especially GM-CSF rescued the induction of GagL85-93-specific CD8+ T cells in mice vaccinated with FV Leader-Gag and Env. Mice that were immunized with plasmids encoding Leader-Gag and Env and the cytokines IL1ß, IL12, IL15, IL28A or GM-CSF, but not Leader-Gag and Env without any cytokine, showed significantly reduced viral loads upon a high-dose Friend virus challenge infection. CONCLUSIONS: Our data demonstrate the potency of cytokine-encoding vectors as adjuvants and immune modulators in composite vaccines for anti-retroviral immunization.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/genética , Virus de la Leucemia Murina de Friend/inmunología , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos , Animales , Citocinas/inmunología , Femenino , Virus de la Leucemia Murina de Friend/genética , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Vectores Genéticos , Inmunización , Inmunomodulación , Interleucina-15/genética , Interleucina-15/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Ratones , Ratones Endogámicos BALB C , Plásmidos , Infecciones por Retroviridae/inmunología , Vacunas de ADN/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Carga Viral
12.
Retrovirology ; 14(1): 42, 2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835242

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) can suppress T cell responses in several different diseases. Previously these suppressive cells were observed to expand in HIV patients and in a mouse retrovirus model, yet their suppressive effect on virus-specific CD8+ T cells in vitro and in vivo has not been characterized thus far. RESULTS: We used the Friend retrovirus (FV) model to demonstrate that MDSCs expand and become activated during the late phase of acute FV infection. Only the subpopulation of granulocytic MDSCs (gMDSCs) but not monocytic MDSC suppressed virus-specific CD8+ T cell proliferation and function in vitro. gMDSCs expressed arginase 1, high levels of the inhibitory ligand PD-L1 and the ATP dephosphorylating enzyme CD39 on the cell surface upon infection. All three molecules were involved in the suppressive effect of the gMDSCs in vitro. MDSC depletion experiments in FV-infected mice revealed that they restrict virus-specific CD8+ T cell responses and thus affect the immune control of chronic retroviruses in vivo. CONCLUSIONS: Our study demonstrates that MDSCs become activated and expand during the acute phase of retrovirus infection. Their suppressive activity on virus-specific CD8+ T cells may contribute to T cell dysfunction and the development of chronic infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Granulocitos/inmunología , Células Supresoras de Origen Mieloide/inmunología , Infecciones por Retroviridae/inmunología , Animales , Antígenos de Diferenciación/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Proliferación Celular , Granulocitos/metabolismo , Granulocitos/patología , Leucemia Experimental/inmunología , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología
13.
Immunity ; 29(5): 782-94, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-19006695

RESUMEN

Chronic viral infections of the hematopoietic system are associated with bone marrow dysfunction, to which both virus-mediated and immune-mediated effects may contribute. Using unresolving noncytopathic Friend virus (FV) infection in mice, we showed that unregulated CD4(+) T cell response to FV caused IFN-gamma-mediated bone marrow pathology and anemia. Importantly, bone marrow pathology was triggered by relative insufficiency in regulatory T (Treg) cells and was prevented by added Treg cells, which suppressed the local IFN-gamma production by FV-specific CD4(+) T cells. We further showed that the T cell receptor (TCR) repertoire of transgenic Treg cells expressing the beta chain of an FV-specific TCR was virtually devoid of FV-specific clones. Moreover, anemia induction by virus-specific CD4(+) T cells was efficiently suppressed by virus-nonspecific Treg cells. Thus, sufficient numbers of polyclonal Treg cells may provide substantial protection against bone marrow pathology in chronic viral infections.


Asunto(s)
Anemia/inmunología , Médula Ósea/inmunología , Médula Ósea/fisiopatología , Linfocitos T CD4-Positivos/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Infecciones por Retroviridae/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Anemia/metabolismo , Anemia/virología , Animales , Médula Ósea/patología , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Crónica , Virus de la Leucemia Murina de Friend/patogenicidad , Técnicas de Silenciamiento del Gen , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Infecciones por Retroviridae/patología , Infecciones por Retroviridae/fisiopatología , Infecciones por Retroviridae/virología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/metabolismo
14.
Retrovirology ; 13: 24, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27076190

RESUMEN

BACKGROUND: Regulatory T cells (Tregs) have been shown to limit anti-viral immunity during chronic retroviral infection and to restrict vaccine-induced T cell responses. The objective of the study was to assess whether a combinational therapy of nanoparticle-based therapeutic vaccination and concomitant transient ablation of Tregs augments anti-viral immunity and improves virus control in chronically retrovirus-infected mice. Therefore, chronically Friend retrovirus (FV)-infected mice were immunized with calcium phosphate (CaP) nanoparticles functionalized with TLR9 ligand CpG and CD8(+) or CD4(+) T cell epitope peptides (GagL85-93 or Env gp70123-141) of FV. In addition, Tregs were ablated during the immunization process. Reactivation of CD4(+) and CD8(+) effector T cells was analysed and the viral loads were determined. RESULTS: Therapeutic vaccination of chronically FV-infected mice with functionalized CaP nanoparticles transiently reactivated cytotoxic CD8(+) T cells and significantly reduced the viral loads. Transient ablation of Tregs during nanoparticle-based therapeutic vaccination strongly enhanced anti-viral immunity and further decreased viral burden. CONCLUSION: Our data illustrate a crucial role for CD4(+) Foxp3(+) Tregs in the suppression of anti-viral T cell responses during therapeutic vaccination against chronic retroviral infection. Thus, the combination of transient Treg ablation and therapeutic nanoparticle-based vaccination confers robust and sustained anti-viral immunity.


Asunto(s)
Leucemia Experimental/terapia , Procedimientos de Reducción del Leucocitos , Nanopartículas/administración & dosificación , Infecciones por Retroviridae/terapia , Linfocitos T Reguladores/inmunología , Infecciones Tumorales por Virus/terapia , Vacunas Virales/administración & dosificación , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Terapia Combinada/métodos , Virus de la Leucemia Murina de Friend/inmunología , Leucemia Experimental/inmunología , Ratones Endogámicos C57BL , Infecciones por Retroviridae/inmunología , Resultado del Tratamiento , Infecciones Tumorales por Virus/inmunología
15.
J Virol ; 89(7): 4011-4, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25589647

RESUMEN

Antiretroviral neutralizing antibody (NAb) responses are often evaluated in the absence of Fc-dependent immune effectors. In murine Friend retrovirus infection, Apobec3/Rfv3 promotes a potent polyclonal NAb response. Here, we show that the Apobec3/Rfv3-dependent NAb response correlated with virus-specific IgG2 titers and that the in vivo neutralization potency of Apobec3/Rfv3-resistant antisera was dependent on activating Fcγ receptors but not complement. The data strengthen retroviral vaccine strategies aimed at eliciting NAbs that activate specific Fcγ receptors.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Citidina Desaminasa/metabolismo , Virus de la Leucemia Murina de Friend/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Animales , Inmunoglobulina G/inmunología , Ratones Endogámicos C57BL
16.
J Virol ; 89(2): 1468-73, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378499

RESUMEN

Toll-like receptor 7 and Myd88 are required for antiretroviral antibody and germinal center responses, but whether somatic hypermutation and class-switch recombination are required for antiretroviral immunity has not been examined. Mice deficient in activation-induced cytidine deaminase (AID) resisted Friend virus infection, produced virus-neutralizing antibodies, and controlled viremia. Passive transfer demonstrated that immune IgM from AID-deficient mice contributes to Friend virus control in the presence of virus-specific CD4+ T cells.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Virus de la Leucemia Murina de Friend/inmunología , Leucemia Experimental/inmunología , Infecciones por Retroviridae/inmunología , Hipermutación Somática de Inmunoglobulina , Infecciones Tumorales por Virus/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citidina Desaminasa/deficiencia , Inmunización Pasiva , Cambio de Clase de Inmunoglobulina , Inmunoglobulina M/inmunología , Leucemia Experimental/virología , Ratones , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología
17.
PLoS Pathog ; 10(3): e1003937, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24651250

RESUMEN

In chronic viral infections, persistent antigen presentation causes progressive exhaustion of virus-specific CD8+ T cells. It has become clear, however, that virus-specific naïve CD8+ T cells newly generated from the thymus can be primed with persisting antigens. In the setting of low antigen density and resolved inflammation, newly primed CD8+ T cells are preferentially recruited into the functional memory pool. Thus, continual recruitment of naïve CD8+ T cells from the thymus is important for preserving the population of functional memory CD8+ T cells in chronically infected animals. Friend virus (FV) is the pathogenic murine retrovirus that establishes chronic infection in adult mice, which is bolstered by the profound exhaustion of virus-specific CD8+ T cells induced during the early phase of infection. Here we show an additional evasion strategy in which FV disseminates efficiently into the thymus, ultimately leading to clonal deletion of thymocytes that are reactive to FV antigens. Owing to the resultant lack of virus-specific recent thymic emigrants, along with the above exhaustion of antigen-experienced peripheral CD8+ T cells, mice chronically infected with FV fail to establish a functional virus-specific CD8+ T cell pool, and are highly susceptible to challenge with tumor cells expressing FV-encoded antigen. However, FV-specific naïve CD8+ T cells generated in uninfected mice can be primed and differentiate into functional memory CD8+ T cells upon their transfer into chronically infected animals. These findings indicate that virus-induced central tolerance that develops during the chronic phase of infection accelerates the accumulation of dysfunctional memory CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica/inmunología , Memoria Inmunológica/inmunología , Infecciones por Retroviridae/inmunología , Timo/virología , Envejecimiento , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Enfermedad Crónica , Femenino , Citometría de Flujo , Virus de la Leucemia Murina de Friend/inmunología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Timo/inmunología
18.
J Immunol ; 193(4): 1567-77, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25000983

RESUMEN

The mechanisms whereby different vaccines may expand distinct Ag-specific T cell clonotypes or induce disparate degrees of protection are incompletely understood. We found that several delivery modes of a model retroviral Ag, including natural infection, preferentially expanded initially rare high-avidity CD4(+) T cell clonotypes, known to mediate protection. In contrast, the same Ag vectored by human adenovirus serotype 5 induced clonotypic expansion irrespective of avidity, eliciting a predominantly low-avidity response. Nonselective clonotypic expansion was caused by relatively weak adenovirus serotype 5-vectored Ag presentation and was reproduced by replication-attenuated retroviral vaccines. Mechanistically, the potency of Ag presentation determined the speed and, consequently, completion of the CD4(+) T cell response. Whereas faster completion retained the initial advantage of high-avidity clonotypes, slower completion permitted uninhibited accumulation of low-avidity clonotypes. These results highlighted the importance of Ag presentation patterns in determining the clonotypic composition of vaccine-induced T cell responses and ultimately the efficacy of vaccination.


Asunto(s)
Afinidad de Anticuerpos/inmunología , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Selección Clonal Mediada por Antígenos/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Productos del Gen env/inmunología , Animales , Presentación de Antígeno/inmunología , Proliferación Celular , Perfilación de la Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/inmunología , Receptores OX40/genética , Vacunas Virales/inmunología
19.
J Immunol ; 193(6): 2952-60, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25098294

RESUMEN

Vß5(+) regulatory T cells (Tregs), which are specific for a mouse endogenous retroviral superantigen, become activated and proliferate in response to Friend virus (FV) infection. We previously reported that FV-induced expansion of this Treg subset was dependent on CD8(+) T cells and TNF-α, but independent of IL-2. We now show that the inflammatory milieu associated with FV infection is not necessary for induction of Vß5(+) Treg expansion. Rather, it is the presence of activated CD8(+) T cells that is critical for their expansion. The data indicate that the mechanism involves signaling between the membrane-bound form of TNF-α on activated CD8(+) T cells and TNFR2 on Tregs. CD8(+) T cells expressing membrane-bound TNF-α but no soluble TNF-α remained competent to induce strong Vß5(+) Treg expansion in vivo. In addition, Vß5(+) Tregs expressing only TNFR2 but no TNFR1 were still responsive to expansion. Finally, treatment of naive mice with soluble TNF-α did not induce Vß5(+) Treg expansion, but treatment with a TNFR2-specific agonist did. These results reveal a new mechanism of intercellular communication between activated CD8(+) T cell effectors and Tregs that results in the activation and expansion of a Treg subset that subsequently suppresses CD8(+) T cell functions.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Proteínas Portadoras/genética , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Leucemia Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Infecciones por Retroviridae/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Infecciones Tumorales por Virus/inmunología
20.
J Immunol ; 193(1): 306-16, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24872193

RESUMEN

Tetherin/BST-2 is a host restriction factor that could directly inhibit retroviral particle release by tethering nascent virions to the plasma membrane. However, the immunological impact of Tetherin during retrovirus infection remains unknown. We now show that Tetherin influences antiretroviral cell-mediated immune responses. In contrast to the direct antiviral effects of Tetherin, which are dependent on cell surface expression, the immunomodulatory effects are linked to the endocytosis of the molecule. Mice encoding endocytosis-competent C57BL/6 Tetherin exhibited lower viremia and pathology at 7 d postinfection with Friend retrovirus (FV) compared with mice encoding endocytosis-defective NZW/LacJ Tetherin. Notably, antiretroviral protection correlated with stronger NK cell responses. In addition, Friend retrovirus infection levels were significantly lower in wild-type C57BL/6 mice than in Tetherin knockout mice at 2 wk postinfection, and antiretroviral protection correlated with stronger NK cell and virus-specific CD8+ T cell responses. The results demonstrate that Tetherin acts as a modulator of the cell-mediated immune response against retrovirus infection in vivo.


Asunto(s)
Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Leucemia Murina de Friend/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Retroviridae/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Antígenos CD/genética , Linfocitos T CD8-positivos/patología , Virus de la Leucemia Murina de Friend/genética , Células Asesinas Naturales/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Infecciones por Retroviridae/genética , Infecciones por Retroviridae/patología , Factores de Tiempo , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/patología , Viremia/genética , Viremia/inmunología , Viremia/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA