Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Cell ; 149(5): 979-93, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22608084

RESUMO

All cancers carry somatic mutations. The patterns of mutation in cancer genomes reflect the DNA damage and repair processes to which cancer cells and their precursors have been exposed. To explore these mechanisms further, we generated catalogs of somatic mutation from 21 breast cancers and applied mathematical methods to extract mutational signatures of the underlying processes. Multiple distinct single- and double-nucleotide substitution signatures were discernible. Cancers with BRCA1 or BRCA2 mutations exhibited a characteristic combination of substitution mutation signatures and a distinctive profile of deletions. Complex relationships between somatic mutation prevalence and transcription were detected. A remarkable phenomenon of localized hypermutation, termed "kataegis," was observed. Regions of kataegis differed between cancers but usually colocalized with somatic rearrangements. Base substitutions in these regions were almost exclusively of cytosine at TpC dinucleotides. The mechanisms underlying most of these mutational signatures are unknown. However, a role for the APOBEC family of cytidine deaminases is proposed.


Assuntos
Neoplasias da Mama/genética , Análise Mutacional de DNA , Estudo de Associação Genômica Ampla , Mutação , Desaminase APOBEC-1 , Proteína BRCA2/genética , Citidina Desaminase/metabolismo , Feminino , Genes BRCA1 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos
2.
Mol Cell ; 31(4): 474-484, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18722174

RESUMO

Activation-induced deaminase (AID) deaminates deoxycytidine residues in immunoglobulin genes, triggering antibody diversification. Here, by use of two-hybrid and coimmunoprecipitation assays, we identify CTNNBL1 (also known as NAP) as an AID-specific interactor. Mutants of AID that interfere with CTNNBL1 interaction yield severely diminished hypermutation and class switching. Targeted inactivation of CTNNBL1 in DT40 B cells also considerably diminishes IgV diversification. CTNNBL1 is a widely expressed nuclear protein that associates with the Prp19 complex of the spliceosome, interacting with its CDC5L component. The results, therefore, identify residues in AID involved in its in vivo targeting and suggest they might act through interaction with CTNNBL1, giving possible insight into the linkage between AID recruitment and target-gene transcription.


Assuntos
Diversidade de Anticorpos , Proteínas Reguladoras de Apoptose/metabolismo , Citidina Desaminase/metabolismo , Proteínas Nucleares/metabolismo , Spliceossomos/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Galinhas , Humanos , Proteínas Mutantes/metabolismo , Mutação/genética , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Ratos , Técnicas do Sistema de Duplo-Híbrido
3.
Semin Immunol ; 24(4): 273-80, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22698843

RESUMO

Although AID fulfils its physiological function of diversifying antibody genes in the nucleus, most of the AID protein within the cell is found in a complex located in the cytoplasm. In this review, we summarize what is currently known about this cytoplasmic AID complex. Its size has been estimated to lie between 300 and 500kDa (sedimentation coefficient of 10-11S) and it comprises the abundant protein translation elongation factor 1α (eEF1A) as a major stoichiometric component. We speculate on the possible roles of this complex as well as of chaperones known to interact with AID in regulating the cytosolic retention of AID and its controlled release for import into the nucleus.


Assuntos
Citidina Desaminase/metabolismo , Citoplasma/metabolismo , Animais , Citidina Desaminase/imunologia , Citoplasma/imunologia , Humanos , Ligação Proteica , Multimerização Proteica , RNA/metabolismo
4.
Nat Rev Immunol ; 5(2): 171-8, 2005 02.
Artigo em Inglês | MEDLINE | ID: mdl-15688043

RESUMO

Somatic hypermutation of immunoglobulin genes occurs at both C.G pairs and A.T pairs. Mutations at C.G pairs are created by activation-induced deaminase (AID)-catalysed deamination of C residues to U residues. Mutations at A.T pairs are probably produced during patch repair of the AID-generated U.G lesion, but they occur through an unknown mechanism. Here, we compare the popular suggestion of nucleotide mispairing through polymerase error with an alternative possibility, mutation through incorporation of dUTP (or another non-canonical nucleotide).


Assuntos
Pareamento Incorreto de Bases/genética , DNA Polimerase Dirigida por DNA , Nucleotídeos de Desoxiuracil/genética , Hipermutação Somática de Imunoglobulina/genética , Adenina , Animais , Pareamento de Bases/genética , Humanos , Timina
5.
Eur J Immunol ; 44(7): 1925-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24771041

RESUMO

Excision of uracil introduced into the immunoglobulin loci by AID is central to antibody diversification. While predominantly carried out by the UNG uracil-DNA glycosylase as reflected by deficiency in immunoglobulin class switching in Ung(-/-) mice, the deficiency is incomplete, as evidenced by the emergence of switched IgG in the serum of Ung(-/-) mice. Lack of switching in mice deficient in both UNG and MSH2 suggested that mismatch repair initiated a backup pathway. We now show that most of the residual class switching in Ung(-/-) mice depends upon the endogenous SMUG1 uracil-DNA glycosylase, with in vitro switching to IgG1 as well as serum IgG3, IgG2b, and IgA greatly diminished in Ung(-/-) Smug1(-/-) mice, and that Smug1 partially compensates for Ung deficiency over time. Nonetheless, using a highly MSH2-dependent mechanism, Ung(-/-) Smug1(-/-) mice can still produce detectable levels of switched isotypes, especially IgG1. While not affecting the pattern of base substitutions, SMUG1 deficiency in an Ung(-/-) background further reduces somatic hypermutation at A:T base pairs. Our data reveal an essential requirement for uracil excision in class switching and in facilitating noncanonical mismatch repair for the A:T phase of hypermutation presumably by creating nicks near the U:G lesion recognized by MSH2.


Assuntos
Switching de Imunoglobulina , Mutação , Uracila-DNA Glicosidase/fisiologia , Uracila/metabolismo , Animais , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos , Proteína 2 Homóloga a MutS/fisiologia
6.
J Immunol ; 190(4): 1481-90, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23303672

RESUMO

Mice transgenic for human Ig loci are an invaluable resource for the production of human Abs. However, such mice often do not yield human mAbs as effectively as conventional mice yield mouse mAbs. Suboptimal efficacy in delivery of human Abs might reflect imperfect interaction between the human membrane IgH chains and the mouse cellular signaling machinery. To obviate this problem, in this study we generated a humanized rat strain (OmniRat) carrying a chimeric human/rat IgH locus (comprising 22 human V(H)s, all human D and J(H) segments in natural configuration linked to the rat C(H) locus) together with fully human IgL loci (12 Vκs linked to Jκ-Cκ and 16 Vλs linked to Jλ-Cλ). The endogenous Ig loci were silenced using designer zinc finger nucleases. Breeding to homozygosity resulted in a novel transgenic rat line exclusively producing chimeric Abs with human idiotypes. B cell recovery was indistinguishable from wild-type animals, and human V(D)J transcripts were highly diverse. Following immunization, the OmniRat strain performed as efficiently as did normal rats in yielding high-affinity serum IgG. mAbs, comprising fully human variable regions with subnanomolar Ag affinity and carrying extensive somatic mutations, are readily obtainable, similarly to conventional mAbs from normal rats.


Assuntos
Sítios de Ligação de Anticorpos , Deficiência de IgG/genética , Deficiência de IgG/imunologia , Regiões Constantes de Imunoglobulina/genética , Imunoglobulina G/biossíntese , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/genética , Animais , Sítios de Ligação de Anticorpos/genética , Cromossomos Artificiais Bacterianos/genética , Cromossomos Artificiais de Levedura/genética , Homologia de Genes/genética , Células Germinativas/imunologia , Células Germinativas/metabolismo , Humanos , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Camundongos , Camundongos Transgênicos , Ratos , Ratos Transgênicos
7.
Nucleic Acids Res ; 40(13): 6016-25, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22447450

RESUMO

Deamination of cytosine (C), 5-methylcytosine (mC) and 5-hydroxymethylcytosine (hmC) occurs spontaneously in mammalian DNA with several hundred deaminations occurring in each cell every day. The resulting potentially mutagenic mispairs of uracil (U), thymine (T) or 5-hydroxymethyluracil (hmU) with guanine (G) are substrates for repair by various DNA glycosylases. Here, we show that targeted inactivation of the mouse Smug1 DNA glycosylase gene is sufficient to ablate nearly all hmU-DNA excision activity as judged by assay of tissue extracts from knockout mice as well as by the resistance of their embryo fibroblasts to 5-hydroxymethyldeoxyuridine toxicity. Inactivation of Smug1 when combined with inactivation of the Ung uracil-DNA glycosylase gene leads to a loss of nearly all detectable uracil excision activity. Thus, SMUG1 is the dominant glycosylase responsible for hmU-excision in mice as well as the major UNG-backup for U-excision. Both Smug1-knockout and Smug1/Ung-double knockout mice breed normally and remain apparently healthy beyond 1 year of age. However, combined deficiency in SMUG1 and UNG exacerbates the cancer predisposition of Msh2(-/-) mice suggesting that when both base excision and mismatch repair pathways are defective, the mutagenic effects of spontaneous cytosine deamination are sufficient to increase cancer incidence but do not preclude mouse development.


Assuntos
Reparo do DNA , Pentoxil (Uracila)/análogos & derivados , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo , Animais , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fluoruracila/metabolismo , Marcação de Genes , Predisposição Genética para Doença , Longevidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Homóloga a MutS/genética , Neoplasias Experimentais/genética , Pentoxil (Uracila)/metabolismo , Timidina/análogos & derivados , Timidina/toxicidade , beta-Galactosidase/genética
8.
Proc Natl Acad Sci U S A ; 108(45): 18366-71, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22042842

RESUMO

Activation-induced cytidine deaminase (AID) is a B lymphocyte-specific DNA deaminase that acts on the Ig loci to trigger antibody gene diversification. Most AID, however, is retained in the cytoplasm and its nuclear abundance is carefully regulated because off-target action of AID leads to cancer. The nature of the cytosolic AID complex and the mechanisms regulating its release from the cytoplasm and import into the nucleus remain unknown. Here, we show that cytosolic AID in DT40 B cells is part of an 11S complex and, using an endogenously tagged AID protein to avoid overexpression artifacts, that it is bound in good stoichiometry to the translation elongation factor 1 alpha (eEF1A). The AID/eEF1A interaction is recapitulated in transfected cells and depends on the C-terminal domain of eEF1A (which is not responsible for GTP or tRNA binding). The eEF1A interaction is destroyed by mutations in AID that affect its cytosolic retention. These results suggest that eEF1A is a cytosolic retention factor for AID and extend on the multiple moonlighting functions of eEF1A.


Assuntos
Citidina Desaminase/metabolismo , Citoplasma/enzimologia , Fator 1 de Elongação de Peptídeos/metabolismo , Animais , Linhagem Celular , Galinhas , Ligação Proteica
9.
J Exp Med ; 204(1): 7-10, 2007 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-17190841

RESUMO

Somatic hypermutation (SHM) introduces nucleotide substitutions into immunoglobulin variable (Ig V) region genes at all four bases, but the mutations at C/G and A/T pairs are achieved by distinct mechanisms. Mutations at C/G pairs are a direct consequence of the C-->U deamination catalyzed by activation-induced deaminase (AID). Mutations at A/T pairs, however, require a second mutagenic process that occurs during patch repair of the AID-generated U/G mismatch. Several DNA polymerases have been proposed to play a role in SHM, but accumulating evidence indicates that the mutations at A/T are overwhelmingly achieved by recruitment of DNA polymerase eta.


Assuntos
Citidina Desaminase/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Hipermutação Somática de Imunoglobulina , Animais , Pareamento de Bases , DNA/genética , DNA/metabolismo , Humanos , Camundongos , Modelos Genéticos
10.
J Exp Med ; 203(9): 2085-94, 2006 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-16894013

RESUMO

Immunoglobulin (Ig) class switching is initiated by deamination of C-->U within the immunoglobulin heavy chain locus, catalyzed by activation-induced deaminase (AID). In the absence of uracil-DNA glycosylase (UNG) and the homologue of bacterial MutS (MSH)-2 mismatch recognition protein, the resultant U:G lesions are not processed into switching events but are fixed by replication allowing sites of AID-catalyzed deamination to be identified by the resulting C-->T mutations. We find that AID targets cytosines in both donor and acceptor switch regions (S regions) with the deamination domains initiating approximately 150 nucleotides 3' of the I exon start sites and extending over several kilobases (the IgH intronic enhancer is spared). Culturing B cells with interleukin 4 or interferon gamma specifically enhanced deamination around Sgamma1 and Sgamma2a, respectively. Mutation spectra suggest that, in the absence of UNG and MSH2, AID may occasionally act at the mu switch region in an apparently processive manner, but there is no marked preference for targeting of the transcribed versus nontranscribed strand (even in areas capable of R loop formation). The data are consistent with switch recombination being triggered by transcription-associated, strand-symmetric AID-mediated deamination at both donor and acceptor S regions with cytokines directing isotype specificity by potentiating AID recruitment to the relevant acceptor S region.


Assuntos
Citidina Desaminase/metabolismo , Região de Troca de Imunoglobulinas , Proteína 2 Homóloga a MutS/metabolismo , Uracila-DNA Glicosidase/metabolismo , Animais , Linfócitos B/fisiologia , Citidina Desaminase/genética , Isotipos de Imunoglobulinas , Interferon gama/metabolismo , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Dados de Sequência Molecular , Proteína 2 Homóloga a MutS/genética , Mutação , Hipermutação Somática de Imunoglobulina , Uracila-DNA Glicosidase/genética
11.
Eur J Immunol ; 41(2): 485-90, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268017

RESUMO

Activation-induced deaminase (AID) is a B lymphocyte-specific DNA deaminase that triggers Ig class-switch recombination (CSR) and somatic hypermutation. It shuttles between cytoplasm and nucleus, containing a nuclear export sequence (NES) at its carboxyterminus. Intriguingly, the precise nature of this NES is critical to AID's function in CSR, though not in somatic hypermutation. Many alterations to the NES, while preserving its nuclear export function, destroy CSR ability. We have previously speculated that AID's ability to potentiate CSR may critically depend on the affinity of interaction between its NES and Crm1 exportin. Here, however, by comparing multiple AID NES mutants, we find that - beyond a requirement for threshold Crm1 binding - there is little correlation between CSR and Crm1 binding affinity. The results suggest that CSR, as well as the stabilisation of AID, depend on an interaction between the AID C-terminal decapeptide and factor(s) additional to Crm1.


Assuntos
Citidina Desaminase/metabolismo , Switching de Imunoglobulina/fisiologia , Carioferinas/metabolismo , Sinais de Exportação Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Substituição de Aminoácidos/fisiologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Citidina Desaminase/genética , Estabilidade Enzimática/fisiologia , Humanos , Carioferinas/genética , Camundongos , Camundongos Knockout , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Exportina 1
12.
FASEB J ; 25(4): 1123-32, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21454370

RESUMO

The origin of antibody diversity has intrigued scientists for nearly a century. We now know that the diversity is achieved through a 2-stage process. Gene rearrangement (catalyzed by the RAG1/2 recombinase) allows the production of a primary repertoire of antibodies; targeted deamination of cytosines within these rearranged antibody genes (catalyzed by the DNA deaminase AID) then allows them to be further diversified and matured by somatic hypermutation, gene conversion, and class-switch recombination. Here we review the history of the uncovering of some of these processes, contrasting the relative importance of hypothesis and methodological developments in driving the research at different periods of the work.


Assuntos
Anticorpos/genética , Diversidade de Anticorpos/imunologia , Animais , Linfócitos B/imunologia , Citidina Desaminase/metabolismo , Rearranjo Gênico/fisiologia , Genes de Imunoglobulinas , Proteínas de Homeodomínio/fisiologia , Humanos , Imunoglobulinas/genética , Modelos Imunológicos , Hipermutação Somática de Imunoglobulina/fisiologia
14.
Proc Natl Acad Sci U S A ; 106(16): 6736-41, 2009 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-19351893

RESUMO

The carboxyterminal region of activation-induced deaminase (AID) is required for its function in Ig class switch recombination (CSR) and also contains a nuclear-export sequence (NES). Here, based on an extensive fine-structure mutation analysis of the AID NES, as well as from AID chimeras bearing heterologous NESs, we show that while a functional NES is indeed essential for CSR, it is not sufficient. The precise nature of the NES is critical both for AID stabilization and CSR function: minor changes in the NES can perturb stabilization and CSR without jeopardizing nuclear export. The results indicate that the AID NES fulfills a function beyond simply providing a signal for nuclear export and suggest the possibility that the quality of exportin-binding may be critical to the stabilization of AID and its activity in CSR.


Assuntos
Citidina Desaminase/química , Citidina Desaminase/metabolismo , Switching de Imunoglobulina/imunologia , Sinais de Exportação Nuclear , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Linhagem Celular , Galinhas , Estabilidade Enzimática , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese , Proteínas Mutantes/metabolismo , Mutação Puntual/genética , Transporte Proteico , Relação Estrutura-Atividade
15.
J Biol Chem ; 285(10): 7111-8, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20022958

RESUMO

The apoB RNA-editing enzyme, catalytic polypeptide-like (APOBEC) family of proteins includes APOBEC1, APOBEC3, and activation-induced deaminase, all of which are zinc-dependent cytidine deaminases active on polynucleotides and involved in RNA editing or DNA mutation. In contrast, the biochemical and physiological functions of APOBEC2, a muscle-specific member of the family, are unknown, although it has been speculated, like APOBEC1, to be an RNA-editing enzyme. Here, we show that, although expressed widely in striated muscle (with levels peaking late during myoblast differentiation), APOBEC2 is preferentially associated with slow-twitch muscle, with its abundance being considerably greater in soleus compared with gastrocnemius muscle and, within soleus muscle, in slow as opposed to fast muscle fibers. Its abundance also decreases following muscle denervation. We further show that APOBEC2-deficient mice harbor a markedly increased ratio of slow to fast fibers in soleus muscle and exhibit an approximately 15-20% reduction in body mass from birth onwards, with elderly mutant animals revealing clear histological evidence of a mild myopathy. Thus, APOBEC2 is essential for normal muscle development and maintenance of fiber-type ratios; although its molecular function remains to be identified, biochemical analyses do not especially argue for any role in RNA editing.


Assuntos
Peso Corporal , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Lenta/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Doenças Musculares/genética , Desaminases APOBEC , Animais , Linhagem Celular , Citidina Desaminase/química , Denervação , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Fibras Musculares de Contração Rápida/citologia , Fibras Musculares de Contração Lenta/citologia , Proteínas Musculares/química , Músculo Esquelético/citologia , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia , Multimerização Proteica
16.
J Virol ; 83(22): 11550-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19726503

RESUMO

APOBEC3 proteins are potent restriction factors against retroviral infection in primates. This restriction is accompanied by hypermutations in the retroviral genome that are attributable to the cytidine deaminase activity of the APOBEC3 proteins. Studies of nucleotide sequence diversity among endogenous gammaretroviruses suggest that the evolution of endogenous retroelements could have been shaped by the mutagenic cytidine deaminase activity of APOBEC3. In mice, however, APOBEC3 appears to restrict exogenous murine retroviruses in the absence of detectable levels of deamination. AKV is an endogenous retrovirus that is involved in causing a high incidence of thymic lymphoma in AKR mice. A comparative analysis of several mouse strains revealed a relatively low level of APOBEC3 expression in AKR mice. Here we show that endogenous mouse APOBEC3 restricts AKV infection and that this restriction likely reflects polymorphisms affecting APOBEC3 abundance rather than differences in the APOBEC3 isoforms expressed. We also observe that restriction of AKV by APOBEC3 is accompanied by G-->A hypermutations in the viral genome. Our findings demonstrate that APOBEC3 acts as a restriction factor in rodents affecting the strain tropism of AKV, and they provide good support for the proposal that APOBEC3-mediated hypermutation contributed to the evolution of endogenous rodent retroviral genomes.


Assuntos
Citidina Desaminase/fisiologia , Vírus da Leucemia Murina/fisiologia , Células 3T3 , Animais , Vírus da Leucemia Murina/genética , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Análise de Sequência de Proteína , Infecções Tumorais por Vírus/virologia
17.
J Virol ; 82(9): 4660-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18272574

RESUMO

APOBEC3 proteins are mammal-specific cytidine deaminases that can restrict retroviral infection. The exact mechanism of the restriction remains unresolved, but one model envisions that uracilated retroviral cDNA, generated by cytidine deamination, is the target of cellular glycosylases. While restriction is unaffected by UNG deficiency, it has been suggested that the SMUG1 glycosylase might provide a backup. We found that retroviral restriction can be achieved by introducing human APOBEC3G into chicken cells (consistent with the components necessary for APOBEC3-mediated restriction predating mammalian evolution) and used this assay to show that APOBEC3G-mediated restriction can occur in cells deficient in both UNG and SMUG1.


Assuntos
Citidina Desaminase/farmacologia , HIV-1/efeitos dos fármacos , Infecções por Retroviridae/prevenção & controle , Uracila-DNA Glicosidase/metabolismo , Desaminase APOBEC-3G , Animais , Galinhas , Citidina Desaminase/genética , DNA Viral/metabolismo , Humanos
18.
J Virol ; 82(22): 10998-1008, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18786991

RESUMO

Several members of the apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like complex 3 (APOBEC3) family in primates act as potent inhibitors of retroviral replication. However, lentiviruses have evolved mechanisms to specifically evade host APOBEC3. Likewise, murine leukemia viruses (MuLV) exclude mouse APOBEC3 from the virions and cleave virion-incorporated APOBEC3. Although the betaretrovirus mouse mammary tumor virus has been shown to be susceptible to mouse APOBEC3, it is not known if APOBEC3 has a physiological role in restricting more widely distributed and long-coevolved mouse gammaretroviruses. The pathogenicity of Friend MuLV (F-MuLV) is influenced by several host genes: some directly restrict the cell entry or integration of the virus, while others influence the host immune responses. Among the latter, the Rfv3 gene has been mapped to chromosome 15 in the vicinity of the APOBEC3 locus. Here we have shown that polymorphisms at the mouse APOBEC3 locus indeed influence F-MuLV replication and pathogenesis: the APOBEC3 alleles of F-MuLV-resistant C57BL/6 and -susceptible BALB/c mice differ in their sequences and expression levels in the hematopoietic tissues and in their abilities to restrict F-MuLV replication both in vitro and in vivo. Furthermore, upon infection with the pathogenic Friend virus complex, (BALB/c x C57BL/6)F(1) mice displayed an exacerbated erythroid cell proliferation when the mice carried a targeted disruption of the C57BL/6-derived APOBEC3 allele. These results indicate, for the first time, that mouse APOBEC3 is a physiologically functioning restriction factor to mouse gammaretroviruses.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Sequência de Aminoácidos , Animais , Linhagem Celular , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Leucemia Experimental/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutagênese Insercional , Polimorfismo Genético , Infecções por Retroviridae/imunologia , Alinhamento de Sequência , Infecções Tumorais por Vírus/imunologia , Replicação Viral/imunologia
19.
Adv Immunol ; 94: 37-73, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17560271

RESUMO

The activation-induced cytidine deaminase (AID)/apolipoprotein B RNA-editing catalytic component (APOBEC) family is a vertebrate-restricted subgrouping of a superfamily of zinc (Zn)-dependent deaminases that has members distributed throughout the biological world. AID and APOBEC2 are the oldest family members with APOBEC1 and the APOBEC3s being later arrivals restricted to placental mammals. Many AID/APOBEC family members exhibit cytidine deaminase activity on polynucleotides, although in different physiological contexts. Here, we examine the AID/APOBEC proteins in the context of the entire Zn-dependent deaminase superfamily. On the basis of secondary structure predictions, we propose that the cytosine and tRNA deaminases are likely to provide better structural paradigms for the AID/APOBEC family than do the cytidine deaminases, to which they have conventionally been compared. These comparisons yield predictions concerning likely polynucleotide-interacting residues in AID/APOBEC3s, predictions that are supported by mutagenesis studies. We also focus on a specific comparison between AID and the APOBEC3s. Both are DNA deaminases that function in immunity and are responsible for the hypermutation of their target substrates. AID functions in the adaptive immune system to diversify antibodies with targeted DNA deamination being central to this function. APOBEC3s function as part of an innate pathway of immunity to retroviruses with targeted DNA deamination being central to their activity in retroviral hypermutation. However, the mechanism by which the APOBEC3s fulfill their function of retroviral restriction remains unresolved.


Assuntos
Citidina Desaminase/imunologia , Animais , Citidina Desaminase/química , Citidina Desaminase/fisiologia , DNA/metabolismo , Desaminação , Humanos , Edição de RNA
20.
Trends Biochem Sci ; 28(6): 305-12, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12826402

RESUMO

Functional antibody genes assembled by V(D)J joining are subsequently diversified by somatic hypermutation, gene conversion and class-switch recombination. Recent evidence indicates that all three processes are caused by the deamination of cytosine to uracil at sites within the immunoglobulin (Ig) loci, with the pattern of diversification depending on the pathway used for resolving the initiating dU-dG lesion. Whereas DNA deamination targeted to the endogenous Ig locus triggers a program of somatic gene diversification that underpins adaptive immunity, deamination targeted to foreign DNA might have arisen initially as a form of innate immunity. Furthermore, the observation that members of the DNA deaminase family can target inappropriate genes suggests they might also contribute to mutations during genome evolution, as well as in cancer.


Assuntos
DNA/metabolismo , Genes de Imunoglobulinas , Imunidade , Animais , Desaminação , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa