Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 111(21): 7759-64, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24821801

RESUMO

Somatic hypermutation (SHM) is an integral process in the development of high-affinity antibodies that are important for recovery from viral infections and vaccine-induced protection. Ig SHM occurs predominantly in germinal centers (GC) via the enzymatic activity of activation-induced deaminase (AID). In contrast, the evolutionarily related apolipoprotein B mRNA-editing enzyme, catalytic polypeptide 3 (APOBEC3) proteins are known to restrict retroviruses, including HIV-1. We previously reported that mouse APOBEC3 encodes Recovery from Friend virus 3 (Rfv3), a classical resistance gene in mice that promotes the neutralizing antibody response against retrovirus infection. We now show that APOBEC3/Rfv3 complements AID in driving Ig SHM during retrovirus infection. Analysis of antibody sequences from retrovirus-specific hybridomas and GC B cells from infected mice revealed Ig heavy-chain V genes with significantly increased C-to-T and G-to-A transitions in wild-type as compared with APOBEC3-defective mice. The context of the mutations was consistent with APOBEC3 but not AID mutational activity. These findings help explain the role of APOBEC3/Rfv3 in promoting the neutralizing antibody responses essential for recovery from retroviral infection and highlight APOBEC3-mediated deamination as a previously unidentified mechanism for antibody diversification in vivo.


Assuntos
Anticorpos Neutralizantes/genética , Anticorpos Antivirais/genética , Citidina Desaminase/imunologia , Infecções por Retroviridae/genética , Hipermutação Somática de Imunoglobulina/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Sequência de Bases , Citidina Desaminase/genética , Centro Germinativo/citologia , Camundongos , Dados de Sequência Molecular , Análise de Sequência de DNA , Hipermutação Somática de Imunoglobulina/genética , Baço/citologia
2.
PLoS One ; 8(3): e60500, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23533681

RESUMO

Understanding the host genetics of the immune response in retrovirus infection models could provide insights for basic HIV vaccine discovery. In Friend retrovirus (FV) infection of mice, Fv1 differentially inhibits N-tropic versus B-tropic FV infection by mediating a capsid-dependent post-entry block, Fv2 susceptibility governs splenomegaly induction, and Rfv3 resistance primes a stronger neutralizing antibody response due to more potent Apobec3 activity. Apobec3 polymorphisms in inbred mouse strains correlate with Rfv3 resistance and susceptibility, with one unresolved exception. The 129/OlaHsd (129P2) mouse strain is Fv2 and Rfv3 susceptible based on genotyping, but infection of 129P2 mice with B-tropic FV resulted in strong neutralizing antibody responses and no splenomegaly. Here we confirm that 129P2 mice are Fv1(nr/nr), explaining its resistance to B-tropic FV. Infection of 129P2 mice with NB-tropic FV, which can efficiently infect mice independent of Fv1 genotype, resulted in severe splenomegaly, high levels of viremia and weak neutralizing antibody responses regardless of Apobec3 status. Notably, high-dose B-tropic FV infection of 129P2 Apobec3-deficient mice induced significant adaptive immune responses and conferred high levels of protection following challenge with pathogenic NB-tropic FV. This immunological protection complemented previous studies that N-tropic FV can act as a live-attenuated vaccine in Fv1 (b/b) mice. Altogether, the results obtained in 129P2 mice strengthen the conclusion that Rfv3 is encoded by Apobec3, and highlight Fv1 incompatibility as a retroviral vaccine paradigm in mice. Due to its susceptibility to disease that allows for pathogenic challenge studies, B-tropic FV infection of 129P2 mice may be a useful model to study the immunological pathways induced by retroviral capsid restriction.


Assuntos
Citidina Desaminase/deficiência , Proteínas/metabolismo , Animais , Linhagem Celular , Citometria de Fluxo , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas/genética , Infecções por Retroviridae/imunologia
3.
J Immunol ; 190(4): 1583-90, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23315078

RESUMO

Therapeutic administration of IFN-α in clinical trials significantly reduced HIV-1 plasma viral load and human T-lymphotropic virus type I proviral load in infected patients. The mechanism may involve the concerted action of multiple antiretroviral effectors collectively known as "restriction factors," which could vary in relative importance according to the magnitude of transcriptional induction. However, direct genetic approaches to identify the relevant IFN-α restriction factors will not be feasible in humans in vivo. Meanwhile, mice encode an analogous set of restriction factor genes and could be used to obtain insights on how IFN-α could inhibit retroviruses in vivo. As expected, IFN-α treatment of mice significantly upregulated the transcription of multiple restriction factors including Tetherin/BST2, SAMHD1, Viperin, ISG15, OAS1, and IFITM3. However, a dominant antiretroviral factor, Apobec3, was only minimally induced. To determine whether Apobec3 was necessary for direct IFN-α antiretroviral action in vivo, wild-type and Apobec3-deficient mice were infected with Friend retrovirus, then treated with IFN-α. Treatment of infected wild-type mice with IFN-α significantly reduced acute plasma viral load 28-fold, splenic proviral load 5-fold, bone marrow proviral load 14-fold, and infected bone marrow cells 7-fold, but no inhibition was observed in Apobec3-deficient mice. These findings reveal that IFN-α inhibits acute Friend retrovirus infection primarily through the antiviral effector Apobec3 in vivo, demonstrate that transcriptional induction levels did not predict the mechanism of IFN-α-mediated control, and highlight the potential of the human APOBEC3 proteins as therapeutic targets against pathogenic retrovirus infections.


Assuntos
Antivirais/administração & dosagem , Citidina Desaminase/fisiologia , Vírus da Leucemia Murina de Friend/imunologia , Interferon-alfa/administração & dosagem , Infecções por Retroviridae/terapia , Infecções por Retroviridae/virologia , Replicação Viral/imunologia , Doença Aguda , Animais , Antivirais/uso terapêutico , Células Cultivadas , Citidina Desaminase/deficiência , Citidina Desaminase/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Humanos , Interferon-alfa/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Retroviridae/imunologia , Viremia/imunologia , Viremia/terapia , Viremia/virologia
4.
PLoS Pathog ; 8(3): e1002596, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22457621

RESUMO

Tetherin is a membrane protein of unusual topology expressed from rodents to humans that accumulates enveloped virus particles on the surface of infected cells. However, whether this 'tethering' activity promotes or restricts retroviral spread during acute retrovirus infection in vivo is controversial. We report here the identification of a single nucleotide polymorphism in the Tetherin gene of NZW/LacJ (NZW) mice that mutated the canonical ATG start site to GTG. Translation of NZW Tetherin from downstream ATGs deleted a conserved dual-tyrosine endosomal sorting motif, resulting in higher cell surface expression and more potent inhibition of Friend retrovirus release compared to C57BL/6 (B6) Tetherin in vitro. Analysis of (B6×NZW)F(1) hybrid mice revealed that increased Tetherin cell surface expression in NZW mice is a recessive trait in vivo. Using a classical genetic backcrossing approach, NZW Tetherin expression strongly correlated with decreased Friend retrovirus replication and pathogenesis. However, the protective effect of NZW Tetherin was not observed in the context of B6 Apobec3/Rfv3 resistance. These findings identify the first functional Tetherin polymorphism within a mammalian host, demonstrate that Tetherin cell surface expression is a key parameter for retroviral restriction, and suggest the existence of a restriction factor hierarchy to counteract pathogenic retrovirus infections in vivo.


Assuntos
Antígenos CD/genética , Retrovirus Endógenos/imunologia , Glicoproteínas de Membrana/genética , Polimorfismo de Nucleotídeo Único , Infecções por Retroviridae/imunologia , Animais , Antígenos CD/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Endogamia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos , Infecções por Retroviridae/metabolismo , Infecções por Retroviridae/virologia , Carga Viral , Replicação Viral
5.
PLoS Pathog ; 7(10): e1002284, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21998583

RESUMO

Members of the APOBEC3 family of deoxycytidine deaminases counteract a broad range of retroviruses in vitro through an indirect mechanism that requires virion incorporation and inhibition of reverse transcription and/or hypermutation of minus strand transcripts in the next target cell. The selective advantage to the host of this indirect restriction mechanism remains unclear, but valuable insights may be gained by studying APOBEC3 function in vivo. Apobec3 was previously shown to encode Rfv3, a classical resistance gene that controls the recovery of mice from pathogenic Friend retrovirus (FV) infection by promoting a more potent neutralizing antibody (NAb) response. The underlying mechanism does not involve a direct effect of Apobec3 on B cell function. Here we show that while Apobec3 decreased titers of infectious virus during acute FV infection, plasma viral RNA loads were maintained, indicating substantial release of noninfectious particles in vivo. The lack of plasma virion infectivity was associated with a significant post-entry block during early reverse transcription rather than G-to-A hypermutation. The Apobec3-dependent NAb response correlated with IgG binding titers against native, but not detergent-lysed virions. These findings indicate that innate Apobec3 restriction promotes NAb responses by maintaining high concentrations of virions with native B cell epitopes, but in the context of low virion infectivity. Finally, Apobec3 restriction was found to be saturable in vivo, since increasing FV inoculum doses resulted in decreased Apobec3 inhibition. By analogy, maximizing the release of noninfectious particles by modulating APOBEC3 expression may improve humoral immunity against pathogenic human retroviral infections.


Assuntos
Anticorpos Neutralizantes/imunologia , Formação de Anticorpos , Citidina Desaminase/metabolismo , Vírus da Leucemia Murina de Friend/patogenicidade , Vírion/patogenicidade , Animais , Antivirais/farmacologia , Citidina Desaminase/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fenótipo , Plasma/virologia , Regiões Promotoras Genéticas , RNA Viral/genética , RNA Viral/metabolismo , Transcrição Reversa , Análise de Sequência de RNA , Carga Viral
6.
J Virol ; 85(1): 189-99, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980520

RESUMO

Rfv3 is an autosomal dominant gene that influences the recovery of resistant mice from Friend retrovirus (FV) infection by limiting viremia and promoting a more potent neutralizing antibody response. We previously reported that Rfv3 is encoded by Apobec3, an innate retrovirus restriction factor. However, it was recently suggested that the Rfv3 susceptible phenotype of high viremia at 28 days postinfection (dpi) was more dominantly controlled by the B-cell-activating factor receptor (BAFF-R), a gene that is linked to but located outside the genetically mapped region containing Rfv3. Although one prototypical Rfv3 susceptible mouse strain, A/WySn, indeed contains a dysfunctional BAFF-R, two other Rfv3 susceptible strains, BALB/c and A.BY, express functional BAFF-R genes, determined on the basis of genotyping and B-cell immunophenotyping. Furthermore, transcomplementation studies in (C57BL/6 [B6] × BALB/c)F(1) and (B6 × A.BY)F(1) mice revealed that the B6 Apobec3 gene significantly influences recovery from FV viremia, cellular infection, and disease at 28 dpi. Finally, the Rfv3 phenotypes of prototypic B6, A.BY, A/WySn, and BALB/c mouse strains correlate with reported Apobec3 mRNA expression levels. Overall, these findings argue against the generality of BAFF-R polymorphisms as a dominant mechanism to explain the Rfv3 recovery phenotype and further strengthen the evidence that Apobec3 encodes Rfv3.


Assuntos
Citidina Desaminase/deficiência , Vírus da Leucemia Murina de Friend/fisiologia , Vírus da Leucemia Murina de Friend/patogenicidade , Replicação Viral , Animais , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Genótipo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Retroviridae , Infecções Tumorais por Vírus
7.
Mol Immunol ; 39(14): 885-97, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12686505

RESUMO

Memory B cells of mice with Ig mu transgenes often carry transgene copies that have moved into the Igh locus via somatic translocation. This phenomenon has been attributed to a selection pressure for somatic hypermutations, which generally are observed at much higher frequencies in translocated copies than in ectopic copies. We tested this idea by immunizing Ig-mu transgenic mice in a manner designed to select B cells that required only one V(H) mutation for a switch in antigenic specificity and recruitment into the memory pool. Despite the minimal mutation requirement, hybridomas carrying somatic translocations to the Igh locus were obtained. Importantly, this occurred despite the fact that translocated and untranslocated mu-transgenes were mutated comparably. Evidently, a strong selection advantage was conferred upon B cells by the somatic translocations. Among the hybridomas, translocated mu-transgenes were active, while ectopic mu-transgenes were uniformly silent. The translocated copy that had conferred an affinity-based selection advantage was expressed at the highest level. Moreover, translocated copies were differentially expressed among hybridoma members, which belonged to a common post-mutational lineage. This suggests that adjustments in transgene expression levels had occurred during memory cell development. These results indicate that, apart from their potential influences on somatic hypermutagenesis and class switch recombination, elements in the Igh locus promote the selection of memory B cells in another way, possibly by regulating the level of Ig expression at various stages of antigen-driven differentiation.


Assuntos
Subpopulações de Linfócitos B/imunologia , Regulação da Expressão Gênica/imunologia , Genes de Imunoglobulinas , Cadeias mu de Imunoglobulina/genética , Memória Imunológica/genética , Transgenes , Animais , Ácido Arsanílico/imunologia , Sequência de Bases , Diferenciação Celular , Linhagem da Célula , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Hemocianinas/imunologia , Hibridomas/imunologia , Switching de Imunoglobulina/genética , Switching de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/genética , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Hipermutação Somática de Imunoglobulina/genética , Hipermutação Somática de Imunoglobulina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA