Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 386
Filtrar
1.
PLoS Pathog ; 15(9): e1008043, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31568492

RESUMO

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.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Muromegalovirus/imunologia , Vacinas Virais/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Feminino , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Produtos do Gene env/genética , Produtos do Gene env/imunologia , Vetores Genéticos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus/genética , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/genética
2.
mBio ; 10(1)2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30782653

RESUMO

B cell follicles of the spleen and lymph nodes are immune privileged sites and serve as sanctuaries for infected CD4+ cells in HIV infection. It is assumed that CD8+ T cell responses promote the establishment of the reservoir, as B cell follicles do not permit CD8+ T cell entry. Here we analyzed the infected cell population in the Friend retrovirus (FV) infection and investigated whether FV can similarly infect follicular cells. For analysis of FV-infected cells, we constructed a recombinant FV encoding the bright fluorescent protein mWasabi and performed flow cytometry with cells isolated from spleens, lymph nodes and bone marrow of FV-mWasabi-infected mice. Using t-stochastic neighbor embedding for data exploration, we demonstrate how the target cell population changes during the course of infection. While FV was widely distributed in erythrocytes, myeloid cells, B cells, and CD4+ T cells in the acute phase of infection, the bulk viral load in the late phase was carried by macrophages and follicular B and CD4+ T cells, suggesting that FV persists in cells that are protected from CD8+ T cell killing. Importantly, seeding into follicular cells was equally observed in CD8+ T cell-depleted mice and in highly FV-susceptible mice that mount a very weak immune response, demonstrating that infection of follicular cells is not driven by immune pressure. Our data demonstrate that infection of cells in the B cell follicle is a characteristic of the FV infection, making this murine retrovirus an even more valuable model for development of retrovirus immunotherapy approaches.IMPORTANCE Human immunodeficiency virus is notorious for its ability to avoid clearance by therapeutic interventions, which is partly attributed to the establishment of reservoirs in latently infected cells and cells that reside in immunologically privileged B cell follicles. In the work presented here, we show that cells of the B cell follicle are equally infected by a simple mouse gammaretrovirus. Using fluorescently labeled Friend retrovirus, we found that B cells and T cells in the B cell follicle, while not carrying the bulk of the virus load, were indeed infected by Friend virus in the early acute phase of the infection and persisted in the chronic infection. Our results suggest that infection of follicular cells may be a shared property of lymphotropic viruses and propose the FV infection of mice as a useful model to study strategies for follicular reservoir elimination.


Assuntos
Linfócitos B/virologia , Vírus da Leucemia Murina de Friend/crescimento & desenvolvimento , Leucemia Experimental/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Citometria de Fluxo , Vírus da Leucemia Murina de Friend/genética , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Linfonodos/virologia , Camundongos , Baço/virologia , Coloração e Rotulagem
3.
J Virol ; 91(20)2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28768877

RESUMO

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.


Assuntos
Adenoviridae/genética , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Transgenes , Adenoviridae/imunologia , Vacinas contra Adenovirus/imunologia , Animais , Linfócitos T CD8-Positivos/química , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/imunologia , Vetores Genéticos , Imunização , Interleucina-2/administração & dosagem , Interleucina-2/genética , Interleucina-2/imunologia , Ativação Linfocitária , Camundongos , Retroviridae/genética , Retroviridae/imunologia , Vacinas de DNA/imunologia
4.
Int J Oncol ; 51(2): 456-466, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28586009

RESUMO

The ETS transcription factors play a critical role during hematopoiesis. In F-MuLV-induced erythroleukemia, Fli­1 insertional activation producing high expression of this transcription factor required to promote proliferation. How deregulated Fli­1 expression alters the balance between erythroid differentiation and proliferation is unknown. To address this issue, we exogenously overexpressed Fli­1 in an erythroleukemic cell harboring activation of spi­1/PU.1, another ETS gene involved in erythroleukemogenesis. While the proliferation in culture remains unaffected, Fli­1 overexpression imparts morphological and immunohistochemical characteristics of immature erythroid progenitors. Fli­1 overexpression in erythroleukemic cells increased the numbers of erythroid colonies on methylcellulose and reduced tumorigenicity as evidenced by increase latency of erythroleukemogenesis in mice inoculated with these cells. Although all transplanted mice developed enlargement of the spleen and liver due to leukemic infiltration, Fli­1 overexpression altered the hematopoietic phenotype, significantly increasing the expression of regulatory hematopoietic genes cKIT, SCA-1, CD41 and CD71. In contrast, expression of Spi­1/PU.1 in a Fli­1 producing erythroleukemia cell line in which fli­1 is activated, resulted in increased proliferation through activation of growth promoting proteins MAPK, AKT, cMYC and JAK2. Importantly, these progenitors express high levels of markers such as CD71 and TER119 associated with more mature erythroid cells. Thus, Fli­1 overexpression induces a de-differentiation program by reverting CFU-E to BFU-E erythroid progenitor activity, while Spi­1/PU.1 promoting maturation from BFU-E to CFU-E.


Assuntos
Proteínas de Ligação a DNA/genética , Leucemia Eritroblástica Aguda/genética , Proteínas de Neoplasias/genética , Proteína Proto-Oncogênica c-fli-1/genética , Animais , Diferenciação Celular/genética , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Regulação Neoplásica da Expressão Gênica/genética , Hematopoese/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Leucemia Eritroblástica Aguda/patologia , Leucemia Eritroblástica Aguda/virologia , Camundongos , Peptídeos/genética
5.
Retrovirology ; 14(1): 28, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-28449719

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/genética , Vírus da Leucemia Murina de Friend/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Citocinas/imunologia , Feminino , Vírus da Leucemia Murina de Friend/genética , Produtos do Gene gag/genética , Produtos do Gene gag/imunologia , Vetores Genéticos , Imunização , Imunomodulação , Interleucina-15/genética , Interleucina-15/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos , Infecções por Retroviridae/imunologia , Vacinas de DNA/administração & dosagem , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Carga Viral
6.
Retrovirology ; 14(1): 8, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28166802

RESUMO

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.


Assuntos
Adenoviridae/genética , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Vacinação/métodos , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/imunologia , Vetores Genéticos , Células HEK293 , Humanos , Camundongos , Infecções por Retroviridae/virologia , Vacinas Combinadas/administração & dosagem , Vacinas Combinadas/genética , Vacinas Combinadas/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/genética
7.
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
8.
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
9.
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
10.
J Virol ; 88(18): 10541-55, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24965475

RESUMO

UNLABELLED: B and CD4(+) T lymphocytes are natural targets of murine leukemia virus (MLV). Migrating lymphocytes adopt a polarized morphology with a trailing edge designated the uropod. Here, we demonstrate that MLV Gag localizes to the uropod in polarized B cells and CD4(+) T cells. The uropod localization of MLV Gag was dependent on plasma membrane (PM) association and multimerization of Gag but independent of the viral glycoprotein Env. Basic residues in MA that are required for MLV Gag recruitment to virological synapses between HEK293 and XC cells were dispensable for uropod localization in migrating B cells. Ultrastructural studies indicated that both wild-type and basic-residue mutant Gag localized to the outer surface of the PM at the uropod. Late-domain mutant virus particles were seen at the uropod in form of budding-arrested intermediates. Finally, uropods mediated contact between MLV-infected B cells and uninfected T cells to form virological synapses. Our results suggest that MLV, not unlike HIV, accumulates at the uropod of primary lymphocytes to facilitate viral spreading through the formation of uropod-mediated cell-cell contacts. IMPORTANCE: Viruses have evolved mechanisms to coordinate their assembly and budding with cell polarity to facilitate their spreading. In this study, we demonstrated that the viral determinants for MLV Gag to localize to the uropod in polarized B cells are distinct from the requirements to localize to virological synapses in transformed cell lines. Basic residues in MA that are required for the Gag localization to virological synapses between HEK293 and XC cells are dispensable for Gag localization to the uropod in primary B cells. Rather, plasma membrane association and capsid-driven multimerization of Gag are sufficient to drive MLV Gag to the uropod. MLV-laden uropods also mediate contacts between MLV-infected B cells and uninfected T cells to form virological synapses. Our results indicate that MLV accumulates at the uropod of primary lymphocytes to facilitate viral spreading through the formation of uropod-mediated cell-cell contacts.


Assuntos
Linfócitos B/virologia , Vírus da Leucemia Murina de Friend/metabolismo , Produtos do Gene gag/metabolismo , Infecções por Retroviridae/veterinária , Doenças dos Roedores/virologia , Linfócitos T/virologia , Animais , Linfócitos B/citologia , Membrana Celular/virologia , Movimento Celular , Polaridade Celular , Células Cultivadas , Vírus da Leucemia Murina de Friend/genética , Produtos do Gene gag/genética , Camundongos , Transporte Proteico , Infecções por Retroviridae/fisiopatologia , Infecções por Retroviridae/virologia , Doenças dos Roedores/fisiopatologia , Linfócitos T/citologia
11.
Microbiol Immunol ; 58(8): 474-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24935657

RESUMO

As splicing was previously found to be important for increasing Friend murine leukemia virus env-mRNA stability and translation, we investigated whether splicing of env-mRNA affected the poly(A) tail length using env expression vectors that yielded unspliced or spliced env-mRNA. Incomplete polyadenylation was detected in a fraction of the unspliced env-mRNA products in an env gene-dependent manner, showing that splicing of Friend murine leukemia virus plays an important role in the efficiency of complete polyadenylation of env-mRNA. These results suggested that the promotion of complete polyadenylation of env-mRNA by splicing might partially explain up-regulation of Env protein expression as a result of splicing.


Assuntos
Vírus da Leucemia Murina de Friend/genética , Produtos do Gene env/genética , Splicing de RNA , RNA Mensageiro/metabolismo , Vírus da Leucemia Murina de Friend/metabolismo , Regulação Viral da Expressão Gênica , Produtos do Gene env/metabolismo , Poliadenilação , RNA Mensageiro/genética
12.
J Immunol ; 193(1): 306-16, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24872193

RESUMO

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.


Assuntos
Antígenos CD/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Glicoproteínas de Membrana/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Antígenos CD/genética , Linfócitos T CD8-Positivos/patologia , Vírus da Leucemia Murina de Friend/genética , Células Matadoras Naturais/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Infecções por Retroviridae/genética , Infecções por Retroviridae/patologia , Fatores de Tempo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/patologia , Viremia/genética , Viremia/imunologia , Viremia/patologia
13.
Microbiol Immunol ; 58(1): 38-50, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24236664

RESUMO

The genome of the Friend murine leukemia virus (Fr-MLV) contains a 5' splice site (5'ss) located at 205 nt and a 3'ss located at 5489 nt. In our previous studies, it was shown that if the HindIII-BglII (879-1904 bp) fragment within gag is deleted from the proA8m1 vector, which carries the entire Fr-MLV sequence, then cryptic splicing of env-mRNA occurs. Here, attempts were made to identify the genomic segment(s) in this region that is/are essential to correct splicing. First, vectors with a serially truncated HindIII-BglII fragment were constructed. The vector, in which a 38 bp fragment (1612-1649 bp) is deleted or reversed in proA8m1, only produced splice variants. It was found that a 38 nt region within gag contains important elements that positively regulate splicing at the correct splice sites. Further analyses of a series of vectors carrying the 38 bp fragment and its flanking sequences showed that a region (1183-1611 nt) upstream of the 38 nt fragment also contains sequences that positively or negatively influence splicing at the correct splice sites. The SphI-NdeI (5140-5400 bp) fragment just upstream of the 3'ss was deleted from vectors that carried the 38 bp fragment and its flanking sequences, which yielded correctly spliced mRNA; interestingly, these deleted vectors showed cryptic splicing. These findings suggest that the 5140-5400 nt region located just upstream of the 3'ss is required for the splicing function of the 38 nt fragment and its flanking sequences.


Assuntos
Processamento Alternativo , Vírus da Leucemia Murina de Friend/genética , Genes gag , Sítios de Splice de RNA , Sequências Reguladoras de Ácido Nucleico , Animais , Sequência de Bases , Linhagem Celular , Gammaretrovirus/genética , Ordem dos Genes , Genoma Viral , Camundongos , Provírus/genética , Alinhamento de Sequência
15.
J Virol ; 87(24): 13760-74, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109240

RESUMO

To assess the possible contribution of host immune responses to the exertion of Fv2-associated resistance to Friend virus (FV)-induced disease development, we inoculated C57BL/6 (B6) mice that lacked various subsets of lymphocytes with FV containing no lactate dehydrogenase-elevating virus. Fv2(r) B6 mice lacking CD4(+) T cells developed early polycythemia and fatal erythroleukemia, while B6 mice lacking CD8(+) T cells remained resistant. Erythroid progenitor cells infected with spleen focus-forming virus (SFFV) were eliminated, and no polycythemia was observed in B cell-deficient B6 mice, but they later developed myeloid leukemia associated with oligoclonal integration of ecotropic Friend murine leukemia virus. Additional depletion of natural killer and/or CD8(+) T cells from B cell-deficient B6 mice resulted in the expansion of SFFV proviruses and the development of polycythemia, indicating that SFFV-infected erythroid cells are not only restricted in their growth but are actively eliminated in Fv2(r) mice through cellular immune responses.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Imunidade Celular , Imunidade Humoral , Leucemia Eritroblástica Aguda/veterinária , Doenças dos Roedores/imunologia , Animais , Linfócitos B/imunologia , Progressão da Doença , Resistência à Doença , Feminino , Vírus da Leucemia Murina de Friend/genética , Humanos , Leucemia Eritroblástica Aguda/imunologia , Leucemia Eritroblástica Aguda/virologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Doenças dos Roedores/virologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/imunologia , Linfócitos T/imunologia
16.
J Virol ; 87(23): 12805-13, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24049172

RESUMO

Nonnative viral glycoproteins, including Friend murine leukemia virus envelope (F-MLV Env) are actively recruited to HIV-1 assembly sites by an unknown mechanism. Because interactions with the lipid microenvironment at budding sites could contribute to recruitment, we examined the contribution of the hydrophobicity of the F-MLV Env membrane-spanning domain (MSD) to its incorporation into HIV-1 particles. A series of F-MLV Env mutants that added or deleted one, two, or three leucines in the MSD were constructed. All six mutants retained the ability to be incorporated into HIV-1 particles, but the -1L, -2L, -3L, +1L, and +2L mutants were not capable of producing infectious particles. Surprisingly, the +3L Env glycoprotein was able to produce infectious particles and was constitutively fusogenic. However, when the cytoplasmic tail domains (CTDs) in the Env constructs were deleted, all six of the MSD mutants were able to produce infectious particles. Further mutational analyses revealed that the first 10 amino acids of the CTD is a critical regulator of infectivity. A similar phenotype was observed in HIV-1 Env upon addition of leucines in the MSD, with +1 and +2 leucine mutations greatly reducing Env activity, but +3 leucine mutations behaving similar to the wild type. Unlike F-MLV Env (+1L and +2L), HIV-1 Env (+1L and +2L) infectivity was not restored by deletion of the CTD. We hypothesize that the CTD forms a coiled-coil that disrupts the protein's functionality if it is not in phase with the trimer interface of the ectodomain.


Assuntos
Membrana Celular/virologia , Citoplasma/virologia , Vírus da Leucemia Murina de Friend/metabolismo , HIV-1/fisiologia , Infecções por Retroviridae/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Vírus da Leucemia Murina de Friend/química , Vírus da Leucemia Murina de Friend/genética , HIV-1/química , HIV-1/genética , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Montagem de Vírus
17.
Microbiol Immunol ; 57(8): 594-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23945025

RESUMO

In addition to the env gene, a 0.3-kb fragment containing the R-U5-5' leader sequence is essential for the induction of spongiform neurodegeneration by Friend murine leukemia virus (Fr-MLV) clone A8 and it also influences expression of the Env protein. Kinetic studies were carried out using two recombinant viruses, R7f, carrying the A8 0.3-kb fragment, and Rec5, carrying the 0.3-kb fragment of the non-neuropathogenic Fr-MLV clone 57. These analyses suggested that the 0.3-kb fragment influenced the expression level of the Env protein by regulating the amount of spliced env-mRNA rather than the amount of total viral mRNA or viral production.


Assuntos
Regiões 5' não Traduzidas , Vírus da Leucemia Murina de Friend/genética , Regulação Viral da Expressão Gênica , RNA Viral/genética , Infecções por Retroviridae/veterinária , Doenças dos Roedores/virologia , Animais , Vírus da Leucemia Murina de Friend/química , Vírus da Leucemia Murina de Friend/metabolismo , Produtos do Gene env/química , Produtos do Gene env/genética , Produtos do Gene env/metabolismo , Cinética , Camundongos , Splicing de RNA , RNA Viral/química , RNA Viral/metabolismo , Infecções por Retroviridae/virologia
18.
Int J Oncol ; 43(1): 169-76, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23677281

RESUMO

Identification of retrovirus integration sites is a powerful method to identify cancer-related genes. This approach led to the discovery of the Friend murine leukemia virus (F-MuLV) integration site-1 (fli-1). Viral insertion at the fli-1 locus induces erythroleukemia in susceptible strains of mice. Our recent data demonstrated that, F-MuLV-infected SCID mice, in contrast to wt CB17 controls, developed a non­erythroleukemic leukemia without viral integration at the fli-1 locus. Using ligation-mediated polymerase chain reaction (LM-PCR) approach we identified a total of 15 viral integration sites in F-MuLV-infected SCID mice. One of the identified insertion sites was located about 62 kb upstream of the myeloblastosis (myb) gene. While integration within or surrounding the myb gene has been reported before for murine leukemia viruses, the location of the viral integration site identified in F-MuLV­infected SCID mice is novel and has never been reported. Using PCR analysis we showed that viral integration at the myb locus occurs with a frequency of 35% and therefore is considered as a common integration site. Integration of F-MuLV in this locus resulted in upregulation of the MYB protein. Flow cytometry analysis and methylcellulose culture of leukemic cells isolated from tumors with viral integration close to the myb indicated tumors of myeloid origin. Our findings indicate that, in contrast to wt CB17 mice, F-MuLV-infected SCID mice display viral integration within myeloid specific gene loci that result in the development of myelogenous leukemia.


Assuntos
Vírus da Leucemia Murina de Friend/genética , Leucemia Mieloide/genética , Leucemia Mieloide/virologia , Proteína Proto-Oncogênica c-fli-1/genética , Integração Viral/genética , Animais , Modelos Animais de Doenças , Vírus da Leucemia Murina de Friend/patogenicidade , Humanos , Leucemia Experimental/genética , Leucemia Mieloide/patologia , Camundongos , Camundongos SCID , Mutagênese Insercional/genética , Oncogenes , Proteínas Proto-Oncogênicas c-myb/genética , Transativadores/genética
19.
Genes Immun ; 14(6): 365-72, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23698707

RESUMO

Using an experimental evolution approach, we recently demonstrated that the mouse-specific pathogen Friend virus (FV) complex adapted to specific major histocompatibility complex (MHC) genotypes, which resulted in fitness tradeoffs when viruses were exposed to hosts possessing novel MHC polymorphisms. Here we report the analysis of patterns of pathogen adaptation and virulence evolution from viruses adapting to one of three hosts that differ across the entire genome (A/WySn, DBA/2J and BALB/c). We found that serial passage of FV complex through these mouse genotypes resulted in significant increases in pathogen fitness (156-fold) and virulence (11-fold). Adaptive responses by post-passage viruses also resulted in host-genotype-specific patterns of adaptation. To evaluate the relative importance of MHC versus non-MHC polymorphisms as factors influencing pathogen adaptation and virulence, we compared the magnitude of fitness tradeoffs incurred by post-passage viruses when infecting hosts possessing either novel MHC polymorphisms alone or hosts possessing novel MHC and non-MHC polymorphisms. MHC polymorphisms alone accounted for 71% and 83% of the total observed reductions in viral fitness and virulence in unfamiliar host genotypes, respectively. Strikingly, these data suggest that genetic polymorphisms within the MHC, a gene region representing only -0.1% of the genome, are major host factors influencing pathogen adaptation and virulence evolution.


Assuntos
Adaptação Fisiológica/genética , Evolução Molecular , Vírus da Leucemia Murina de Friend/patogenicidade , Interações Hospedeiro-Parasita , Complexo Principal de Histocompatibilidade/genética , Polimorfismo Genético , Animais , Vírus da Leucemia Murina de Friend/genética , Aptidão Genética , Genótipo , Especificidade de Hospedeiro , Camundongos , Camundongos Endogâmicos , Virulência/genética
20.
Virol J ; 10: 124, 2013 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-23602143

RESUMO

BACKGROUND: A neuropathogenic variant of Friend murine leukemia virus (Fr-MLV) clone A8 induces spongiform neurodegeneration when infected into neonatal rats. Studies with chimeras constructed from the A8 virus and the non-neuropathogenic Fr-MLV clone 57 identified a 0.3-kb KpnI-AatII fragment containing a R-U5-5'leader sequence as an important determinant for inducing spongiosis, in addition to the env gene of A8 as the primary determinant. This 0.3-kb fragment contains a 17-nucleotide difference between the A8 and 57 sequences. We previously showed that the 0.3-kb fragment influences expression levels of Env protein in both cultured cells and rat brain, but the corresponding molecular mechanisms are not well understood. RESULTS: Studies with expression vectors constructed from the full-length proviral genome of Fr-MLV that incorporated the luciferase (luc) gene instead of the env gene found that the vector containing the A8-0.3-kb fragment yielded a larger amount of spliced luc-mRNA and showed higher expression of luciferase when compared to the vector containing the 57-0.3-kb fragment. The amount of total transcripts from the vectors, the poly (A) tail length of their mRNAs, and the nuclear-cytoplasm distribution of luc-mRNA in transfected cells were also evaluated. The 0.3-kb fragment did not influence transcription efficiency, mRNA polyadenylation or nuclear export of luc-mRNA. Mutational analyses were carried out to determine the importance of nucleotides that differ between the A8 and 57 sequences within the 0.3-kb fragment. In particular, seven nucleotides upstream of the 5'splice site (5'ss) were found to be important in regulating the level of protein expression from spliced messages. Interestingly, these nucleotides reside within the stem-loop structure that has been speculated to limit the recognition of 5'ss. CONCLUSIONS: The 0.3-kb fragment containing the R-U5-5'leader sequence of Fr-MLV influences the level of protein expression from the spliced-mRNA by regulating the splicing efficiency rather than transcription, nuclear export of spliced-mRNA, or poly (A) addition to mRNA. Seven nucleotides in the 0.3-kb fragment, which reside within the stem-loop structure that has been speculated to limit recognition of the 5'ss, could pinpoint the function of this region.


Assuntos
Regiões 5' não Traduzidas , Vírus da Leucemia Murina de Friend/genética , Regulação Viral da Expressão Gênica , RNA Mensageiro/genética , RNA Viral/genética , Proteínas do Envelope Viral/biossíntese , Animais , Fusão Gênica Artificial , Genes Reporter , Luciferases/análise , Luciferases/genética , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA