Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Immunol ; 202(5): 1573-1581, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30665938

RESUMO

Antibody diversity is initiated by activation-induced deaminase (AID), which deaminates cytosine to uracil in DNA. Uracils in the Ig gene loci can be recognized by uracil DNA glycosylase (UNG) or mutS homologs 2 and 6 (MSH2-MSH6) proteins, and then processed into DNA breaks. Breaks in switch regions of the H chain locus cause isotype switching and have been extensively characterized as staggered and blunt double-strand breaks. However, breaks in V regions that arise during somatic hypermutation are poorly understood. In this study, we characterize AID-dependent break formation in JH introns from mouse germinal center B cells. We used a ligation-mediated PCR assay to detect single-strand breaks and double-strand breaks that were either staggered or blunt. In contrast to switch regions, V regions contained predominantly single-strand breaks, which peaked 10 d after immunization. We then examined the pathways used to generate these breaks in UNG- and MSH6-deficient mice. Surprisingly, both DNA repair pathways contributed substantially to break formation, and in the absence of both UNG and MSH6, the frequency of breaks was severely reduced. When the breaks were sequenced and mapped, they were widely distributed over a 1000-bp intron region downstream of JH3 and JH4 exons and were unexpectedly located at all 4 nt. These data suggest that during DNA repair, nicks are generated at distal sites from the original deaminated cytosine, and these repair intermediates could generate both faithful and mutagenic repair. During mutagenesis, single-strand breaks would allow entry for low-fidelity DNA polymerases to generate somatic hypermutation.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/genética , Região Variável de Imunoglobulina/genética , Uracila-DNA Glicosidase/genética , Animais , Quebras de DNA , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/imunologia , Região Variável de Imunoglobulina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/imunologia
2.
Free Radic Biol Med ; 131: 59-71, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472364

RESUMO

Uracil-DNA glycosylase (UNG) initiates the base excision repair pathway by excising uracil from DNA. We have previously shown that Trypanosoma brucei cells defective in UNG exhibit reduced infectivity thus demonstrating the relevance of this glycosylase for survival within the mammalian host. In the early steps of the immune response, nitric oxide (NO) is released by phagocytes, which in combination with oxygen radicals produce reactive nitrogen species (RNS). These species can react with DNA generating strand breaks and base modifications including deaminations. Since deaminated cytosines are the main substrate for UNG, we hypothesized that the glycosylase might confer protection towards nitrosative stress. Our work establishes the occurrence of genotoxic damage in Trypanosoma brucei upon exposure to NO in vitro and shows that deficient base excision repair results in increased levels of damage in DNA and a hypermutator phenotype. We also evaluate the incidence of DNA damage during infection in vivo and show that parasites recovered from mice exhibit higher levels of DNA strand breaks, base deamination and repair foci compared to cells cultured in vitro. Notably, the absence of UNG leads to reduced infectivity and enhanced DNA damage also in animal infections. By analysing mRNA and protein levels, we found that surviving UNG-KO trypanosomes highly express tryparedoxin peroxidase involved in trypanothione/tryparedoxin metabolism. These observations suggest that the immune response developed by the host enhances the activation of genes required to counteract oxidative stress and emphasize the importance of DNA repair pathways in the protection to genotoxic and oxidative stress in trypanosomes.


Assuntos
Reparo do DNA , DNA de Protozoário/genética , Óxido Nítrico/farmacologia , Proteínas de Protozoários/genética , Trypanosoma brucei brucei/genética , Uracila-DNA Glicosidase/genética , Animais , Dano ao DNA , DNA de Protozoário/imunologia , Feminino , Expressão Gênica , Genótipo , Glutationa/análogos & derivados , Glutationa/metabolismo , Interações Hospedeiro-Parasita , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Estresse Nitrosativo/genética , Parasitemia/imunologia , Parasitemia/metabolismo , Parasitemia/parasitologia , Peroxidases/genética , Peroxidases/metabolismo , Fenótipo , Proteínas de Protozoários/metabolismo , Espermidina/análogos & derivados , Espermidina/metabolismo , Tiorredoxinas/metabolismo , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/patogenicidade , Tripanossomíase/imunologia , Tripanossomíase/metabolismo , Tripanossomíase/parasitologia , Uracila-DNA Glicosidase/deficiência
4.
Mol Cancer Res ; 16(2): 212-221, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29117941

RESUMO

Thymidylate synthase (TS) inhibitors including fluoropyrimidines [e.g., 5-Fluorouracil (5-FU) and 5-Fluorodeoxyuridine (5-FdU, floxuridine)] and antifolates (e.g., pemetrexed) are widely used against solid tumors. Previously, we reported that shRNA-mediated knockdown (KD) of uracil DNA glycosylase (UDG) sensitized cancer cells to 5-FdU. Because p53 has also been shown as a critical determinant of the sensitivity to TS inhibitors, we further interrogated 5-FdU cytotoxicity after UDG depletion with regard to p53 status. By analyzing a panel of human cancer cells with known p53 status, it was determined that p53-mutated or -deficient cells are highly resistant to 5-FdU. UDG depletion resensitizes 5-FdU in p53-mutant and -deficient cells, whereas p53 wild-type (WT) cells are not affected under similar conditions. Utilizing paired HCT116 p53 WT and p53 knockout (KO) cells, it was shown that loss of p53 improves cell survival after 5-FdU, and UDG depletion only significantly sensitizes p53 KO cells. This sensitization can also be recapitulated by UDG depletion in cells with p53 KD by shRNAs. In addition, sensitization is also observed with pemetrexed in p53 KO cells, but not with 5-FU, most likely due to RNA incorporation. Importantly, in p53 WT cells, the apoptosis pathway induced by 5-FdU is activated independent of UDG status. However, in p53 KO cells, apoptosis is compromised in UDG-expressing cells, but dramatically elevated in UDG-depleted cells. Collectively, these results provide evidence that loss of UDG catalyzes significant cell death signals only in cancer cells mutant or deficient in p53.Implications: This study reveals that UDG depletion restores sensitivity to TS inhibitors and has chemotherapeutic potential in the context of mutant or deficient p53. Mol Cancer Res; 16(2); 212-21. ©2017 AACR.


Assuntos
Desoxiuridina/análogos & derivados , Mutação , Neoplasias/genética , RNA Interferente Pequeno/farmacologia , Proteína Supressora de Tumor p53/genética , Uracila-DNA Glicosidase/deficiência , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desoxiuridina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Técnicas de Silenciamento de Genes/métodos , Técnicas de Inativação de Genes , Células HCT116 , Humanos , Neoplasias/tratamento farmacológico , Pemetrexede/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores
5.
Sci Rep ; 7(1): 7199, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28775312

RESUMO

Both a DNA lesion and an intermediate for antibody maturation, uracil is primarily processed by base excision repair (BER), either initiated by uracil-DNA glycosylase (UNG) or by single-strand selective monofunctional uracil DNA glycosylase (SMUG1). The relative in vivo contributions of each glycosylase remain elusive. To assess the impact of SMUG1 deficiency, we measured uracil and 5-hydroxymethyluracil, another SMUG1 substrate, in Smug1 -/- mice. We found that 5-hydroxymethyluracil accumulated in Smug1 -/- tissues and correlated with 5-hydroxymethylcytosine levels. The highest increase was found in brain, which contained about 26-fold higher genomic 5-hydroxymethyluracil levels than the wild type. Smug1 -/- mice did not accumulate uracil in their genome and Ung -/- mice showed slightly elevated uracil levels. Contrastingly, Ung -/- Smug1 -/- mice showed a synergistic increase in uracil levels with up to 25-fold higher uracil levels than wild type. Whole genome sequencing of UNG/SMUG1-deficient tumours revealed that combined UNG and SMUG1 deficiency leads to the accumulation of mutations, primarily C to T transitions within CpG sequences. This unexpected sequence bias suggests that CpG dinucleotides are intrinsically more mutation prone. In conclusion, we showed that SMUG1 efficiently prevent genomic uracil accumulation, even in the presence of UNG, and identified mutational signatures associated with combined UNG and SMUG1 deficiency.


Assuntos
Citosina/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Uracila-DNA Glicosidase/deficiência , Uracila/metabolismo , Animais , Ilhas de CpG , Desaminação , Genoma , Genômica/métodos , Camundongos , Camundongos Knockout , Mutação
6.
J Exp Med ; 213(11): 2459-2472, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27697833

RESUMO

Activation-induced deaminase (AID) initiates antibody gene diversification by creating G:U mismatches in the immunoglobulin loci. However, AID also deaminates nonimmunoglobulin genes, and failure to faithfully repair these off-target lesions can cause B cell lymphoma. In this study, we identify a mechanism by which processing of G:U produced by AID at the telomeres can eliminate B cells at risk of genomic instability. We show that telomeres are off-target substrates of AID and that B cell proliferation depends on protective repair by uracil-DNA glycosylase (UNG). In contrast, in the absence of UNG activity, deleterious processing by mismatch repair leads to telomere loss and defective cell proliferation. Indeed, we show that UNG deficiency reduces B cell clonal expansion in the germinal center in mice and blocks the proliferation of tumor B cells expressing AID. We propose that AID-induced damage at telomeres acts as a fail-safe mechanism to limit the tumor promoting activity of AID when it overwhelms uracil excision repair.


Assuntos
Linfócitos B/metabolismo , Citidina Desaminase/metabolismo , Citoproteção , Telômero/metabolismo , Uracila-DNA Glicosidase/metabolismo , Animais , Sequência de Bases , Proliferação de Células , Células Clonais , Dano ao DNA/genética , Reparo de Erro de Pareamento de DNA/genética , Centro Germinativo/metabolismo , Switching de Imunoglobulina , Ativação Linfocitária/imunologia , Linfoma de Células B/patologia , Camundongos Endogâmicos C57BL , Modelos Biológicos , Ligação Proteica , Recombinação Genética/genética , Uracila-DNA Glicosidase/deficiência
7.
J Biol Chem ; 291(2): 731-8, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26620559

RESUMO

In mammals, active DNA demethylation involves oxidation of 5-methylcytosine (5mC) into 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) by Tet dioxygenases and excision of these two oxidized bases by thymine DNA glycosylase (TDG). Although TDG is essential for active demethylation in embryonic stem cells and induced pluripotent stem cells, it is hardly expressed in mouse zygotes and dispensable in pronuclear DNA demethylation. To search for other factors that might contribute to demethylation in mammalian cells, we performed a functional genomics screen based on a methylated luciferase reporter assay. UNG2, one of the glycosylases known to excise uracil residues from DNA, was found to reduce DNA methylation, thus activating transcription of a methylation-silenced reporter gene when co-transfected with Tet2 into HEK293T cells. Interestingly, UNG2 could decrease 5caC from the genomic DNA and a reporter plasmid in transfected cells, like TDG. Furthermore, deficiency in Ung partially impaired DNA demethylation in mouse zygotes. Our results suggest that UNG might be involved in Tet-mediated DNA demethylation.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Uracila-DNA Glicosidase/metabolismo , Animais , Citosina/análogos & derivados , DNA/metabolismo , Dioxigenases , Genes Reporter , Loci Gênicos , Genoma Humano , Células HEK293 , Humanos , Camundongos , Plasmídeos/metabolismo , Transfecção , Uracila/metabolismo , Uracila-DNA Glicosidase/deficiência , Zigoto/metabolismo
8.
Behav Brain Res ; 283: 215-26, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25655513

RESUMO

Dietary deficiencies in folic acid result in elevated levels of plasma homocysteine, which has been associated with the development of dementia and other neurodegenerative disorders. Previously, we have shown that elevated levels of plasma homocysteine in mice deficient for a DNA repair enzyme, uracil-DNA glycosylase (UNG), result in neurodegeneration. The goal of this study was to evaluate how deficiencies in folic acid and UNG along with elevated levels of homocysteine affect vascular cognitive impairment, via chronic hypoperfursion in an animal model. Ung(+/+) and Ung(-/-) mice were placed on either control (CD) or folic acid deficient (FADD) diets. Six weeks later, the mice either underwent implantation of microcoils around both common carotid arteries. Post-operatively, behavioral tests began at 3-weeks, angiography was measured after 5-weeks using MRI to assess vasculature and at completion of study plasma and brain tissue was collected for analysis. Learning impairments in the Morris water maze (MWM) were observed only in hypoperfused Ung(-/-) FADD mice and these mice had significantly higher plasma homocysteine concentrations. Interestingly, Ung(+/+) FADD produced significant remodeling of the basilar artery and arterial vasculature. Increased expression of GFAP was observed in the dentate gyrus of Ung(-/-) hypoperfused and FADD sham mice. Chronic hypoperfusion resulted in increased cortical MMP-9 protein levels of FADD hypoperfused mice regardless of genotypes. These results suggest that elevated levels of homocysteine only, as a result of dietary folic acid deficiency, don't lead to memory impairments and neurobiochemical changes. Rather a combination of either chronic hypoperfusion or UNG deficiency is required.


Assuntos
Transtornos Cerebrovasculares/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Deficiência de Ácido Fólico/fisiopatologia , Homocisteína/sangue , Deficiências da Aprendizagem/fisiopatologia , Uracila-DNA Glicosidase/deficiência , Animais , Artéria Basilar/patologia , Artéria Basilar/fisiopatologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Doenças das Artérias Carótidas , Transtornos Cerebrovasculares/patologia , Doença Crônica , Transtornos Cognitivos/patologia , Dieta , Modelos Animais de Doenças , Feminino , Deficiência de Ácido Fólico/patologia , Proteína Glial Fibrilar Ácida , Gliose/patologia , Gliose/fisiopatologia , Deficiências da Aprendizagem/patologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Aprendizagem em Labirinto/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Distribuição Aleatória , Uracila-DNA Glicosidase/genética
9.
J Virol ; 89(6): 3366-79, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589640

RESUMO

UNLABELLED: Uracil DNA glycosylases (UNG) are highly conserved proteins that preserve DNA fidelity by catalyzing the removal of mutagenic uracils. All herpesviruses encode a viral UNG (vUNG), and yet the role of the vUNG in a pathogenic course of gammaherpesvirus infection is not known. First, we demonstrated that the vUNG of murine gammaherpesvirus 68 (MHV68) retains the enzymatic function of host UNG in an in vitro class switch recombination assay. Next, we generated a recombinant MHV68 with a stop codon in ORF46/UNG (ΔUNG) that led to loss of UNG activity in infected cells and a replication defect in primary fibroblasts. Acute replication of MHV68ΔUNG in the lungs of infected mice was reduced 100-fold and was accompanied by a substantial delay in the establishment of splenic latency. Latency was largely, yet not fully, restored by an increase in virus inoculum or by altering the route of infection. MHV68 reactivation from latent splenocytes was not altered in the absence of the vUNG. A survey of host UNG activity in cells and tissues targeted by MHV68 indicated that the lung tissue has a lower level of enzymatic UNG activity than the spleen. Taken together, these results indicate that the vUNG plays a critical role in the replication of MHV68 in tissues with limited host UNG activity and this vUNG-dependent expansion, in turn, influences the kinetics of latency establishment in distal reservoirs. IMPORTANCE: Herpesviruses establish chronic lifelong infections using a strategy of replicative expansion, dissemination to latent reservoirs, and subsequent reactivation for transmission and spread. We examined the role of the viral uracil DNA glycosylase, a protein conserved among all herpesviruses, in replication and latency of murine gammaherpesvirus 68. We report that the viral UNG of this murine pathogen retains catalytic activity and influences replication in culture. The viral UNG was impaired for productive replication in the lung. This defect in expansion at the initial site of acute replication was associated with a substantial delay of latency establishment in the spleen. The levels of host UNG were substantially lower in the lung compared to the spleen, suggesting that herpesviruses encode a viral UNG to compensate for reduced host enzyme levels in some cell types and tissues. These data suggest that intervention at the site of initial replicative expansion can delay the establishment of latency, a hallmark of chronic herpesvirus infection.


Assuntos
Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Rhadinovirus/enzimologia , Doenças dos Roedores/virologia , Uracila-DNA Glicosidase/deficiência , Latência Viral , Replicação Viral , Animais , Feminino , Regulação Viral da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Rhadinovirus/genética , Rhadinovirus/fisiologia , Uracila-DNA Glicosidase/genética
10.
J Biol Chem ; 290(9): 5502-11, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25572391

RESUMO

Uracil in the genome can result from misincorporation of dUTP instead of dTTP during DNA synthesis, and is primarily removed by uracil DNA glycosylase (UNG) during base excision repair. Telomeres contain long arrays of TTAGGG repeats and may be susceptible to uracil misincorporation. Using model telomeric DNA substrates, we showed that the position and number of uracil substitutions of thymine in telomeric DNA decreased recognition by the telomere single-strand binding protein, POT1. In primary mouse hematopoietic cells, uracil was detectable at telomeres, and UNG deficiency further increased uracil loads and led to abnormal telomere lengthening. In UNG-deficient cells, the frequencies of sister chromatid exchange and fragility in telomeres also significantly increased in the absence of telomerase. Thus, accumulation of uracil and/or UNG deficiency interferes with telomere maintenance, thereby underscoring the necessity of UNG-initiated base excision repair for the preservation of telomere integrity.


Assuntos
Células da Medula Óssea/metabolismo , Reparo do DNA , Telômero/metabolismo , Uracila/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Hibridização in Situ Fluorescente , Camundongos Knockout , Ligação Proteica , Complexo Shelterina , Telômero/genética , Homeostase do Telômero/genética , Proteínas de Ligação a Telômeros , Proteína 1 de Ligação a Repetições Teloméricas/genética , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Timina/metabolismo , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética
11.
J Allergy Clin Immunol ; 134(2): 411-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24836470

RESUMO

BACKGROUND: The molecular mechanism of class-switch recombination (CSR) in human subjects has not been fully elucidated. The CSR-induced mutations occurring in the switch region of the IgM gene (Smu-SHMs) in in vitro CSR-activated and in vivo switched B cells have been analyzed in mice but not in human subjects. OBJECTIVE: We sought to better characterize the molecular mechanism of CSR in human subjects. METHODS: Smu-SHMs were analyzed in vitro and in vivo by using healthy control subjects and patients with molecularly defined CSR defects. RESULTS: We found that Smu-SHMs can be induced in vitro by means of CSR activation in human subjects. We also found large amounts of Smu-SHMs in in vivo class-switched memory B cells, smaller (although significant) amounts in unswitched memory B cells, and very low amounts in naive B cells. In class-switched memory B cells a high frequency of Smu-SHMs was found throughout the Smu. In unswitched memory B cells, the Smu-SHM frequency was significantly decreased in the 5' part of the Smu. The difference between switched and unswitched B cells suggests that the extension of somatic hypermutation (SHM) to the 5' upstream region of the Smu might be associated with the effective induction of CSR. The analysis of the pattern of mutations within and outside the WRCY/RGYW (W, A/T; R, A/G; and Y, C/T) motifs, as well as the Smu-SHMs, in CD27(+) B cells from CD40 ligand (CD40L)-, activation-induced cytidine deaminase (AID)-, and uracil-DNA glycosylase (UNG)-deficient patients revealed the dependence of Smu-SHM on CD40L, AID, UNG, and the mismatch repair system in human subjects. CONCLUSION: CD40L-, AID-, UNG-, and mismatch repair system-dependent Smu-SHMs and extension to the 5' region of Smu are necessary to accomplish effective CSR in human subjects.


Assuntos
Ligante de CD40/imunologia , Citidina Desaminase/imunologia , Switching de Imunoglobulina , Imunoglobulina M/genética , Hipermutação Somática de Imunoglobulina , Uracila-DNA Glicosidase/imunologia , Região 5'-Flanqueadora , Adulto , Motivos de Aminoácidos , Linfócitos B/imunologia , Ligante de CD40/deficiência , Ligante de CD40/genética , Criança , Citidina Desaminase/deficiência , Citidina Desaminase/genética , Regulação da Expressão Gênica , Humanos , Imunoglobulina M/imunologia , Memória Imunológica , Lactente , Masculino , Dados de Sequência Molecular , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética
12.
Mol Cell Biol ; 34(12): 2176-87, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24710273

RESUMO

A/T mutations at immunoglobulin loci are introduced by DNA polymerase η (Polη) during an Msh2/6-dependent repair process which results in A's being mutated 2-fold more often than T's. This patch synthesis is initiated by a DNA incision event whose origin is still obscure. We report here the analysis of A/T oligonucleotide mutation substrates inserted at the heavy chain locus, including or not including internal C's or G's. Surprisingly, the template composed of only A's and T's was highly mutated over its entire 90-bp length, with a 2-fold decrease in mutation from the 5' to the 3' end and a constant A/T ratio of 4. These results imply that Polη synthesis was initiated from a break in the 5'-flanking region of the substrate and proceeded over its entire length. The A/T bias was strikingly altered in an Ung(-/-) background, which provides the first experimental evidence supporting a concerted action of Ung and Msh2/6 pathways to generate mutations at A/T bases. New analysis of Pms2(-/-) animals provided a complementary picture, revealing an A/T mutation ratio of 4. We therefore propose that Ung and Pms2 may exert a mutual backup function for the DNA incision that promotes synthesis by Polη, each with a distinct strand bias.


Assuntos
Sequência Rica em At/genética , DNA Glicosilases/deficiência , DNA Glicosilases/metabolismo , DNA/genética , Oligonucleotídeos/genética , Hipermutação Somática de Imunoglobulina/genética , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/metabolismo , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , DNA Glicosilases/genética , Reparo de Erro de Pareamento de DNA , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Técnicas de Introdução de Genes , Loci Gênicos/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Endonuclease PMS2 de Reparo de Erro de Pareamento , Dados de Sequência Molecular , Mutagênese , Mutação/genética , Taxa de Mutação , Especificidade por Substrato , Transgenes/genética , Uracila-DNA Glicosidase/genética
13.
Cell Death Dis ; 5: e1045, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24503537

RESUMO

Misincorporation of genomic uracil and formation of DNA double strand breaks (DSBs) are known consequences of exposure to TS inhibitors such as pemetrexed. Uracil DNA glycosylase (UNG) catalyzes the excision of uracil from DNA and initiates DNA base excision repair (BER). To better define the relationship between UNG activity and pemetrexed anticancer activity, we have investigated DNA damage, DSB formation, DSB repair capacity, and replication fork stability in UNG(+/+) and UNG(-/-) cells. We report that despite identical growth rates and DSB repair capacities, UNG(-/-) cells accumulated significantly greater uracil and DSBs compared with UNG(+/+) cells when exposed to pemetrexed. ChIP-seq analysis of γ-H2AX enrichment confirmed fewer DSBs in UNG(+/+) cells. Furthermore, DSBs in UNG(+/+) and UNG(-/-) cells occur at distinct genomic loci, supporting differential mechanisms of DSB formation in UNG-competent and UNG-deficient cells. UNG(-/-) cells also showed increased evidence of replication fork instability (PCNA dispersal) when exposed to pemetrexed. Thymidine co-treatment rescues S-phase arrest in both UNG(+/+) and UNG(-/-) cells treated with IC50-level pemetrexed. However, following pemetrexed exposure, UNG(-/-) but not UNG(+/+) cells are refractory to thymidine rescue, suggesting that deficient uracil excision rather than dTTP depletion is the barrier to cell cycle progression in UNG(-/-) cells. Based on these findings we propose that pemetrexed-induced uracil misincorporation is genotoxic, contributing to replication fork instability, DSB formation and ultimately cell death.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Glutamatos/farmacologia , Guanina/análogos & derivados , Neoplasias/enzimologia , Neoplasias/genética , Uracila-DNA Glicosidase/deficiência , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Reparo do DNA , Guanina/farmacologia , Humanos , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Pemetrexede , Uracila/metabolismo , Uracila-DNA Glicosidase/genética
14.
Breast Cancer Res Treat ; 142(3): 515-27, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24253812

RESUMO

Uracil in DNA is an important cause of mutagenesis. SMUG1 is a uracil-DNA glycosylase that removes uracil through base excision repair. SMUG1 also processes radiation-induced oxidative base damage as well as 5-fluorouracil incorporated into DNA during chemotherapy. We investigated SMUG1 mRNA expression in 249 primary breast cancers. SMUG1 protein expression was investigated in 1,165 breast tumours randomised into two cohorts [training set (n = 583) and test set (n = 582)]. SMUG1 and chemotherapy response was also investigated in a series of 315 ER-negative tumours (n = 315). For mechanistic insights, SMUG1 was correlated to biomarkers of aggressive phenotype, DNA repair, cell cycle and apoptosis. Low SMUG1 mRNA expression was associated with adverse disease specific survival (p = 0.008) and disease-free survival (p = 0.008). Low SMUG1 protein expression (25 %) was associated with high histological grade (p < 0.0001), high mitotic index (p < 0.0001), pleomorphism (p < 0.0001), glandular de-differentiation (p = 0.0001), absence of hormonal receptors (ER-/PgR-/AR) (p < 0.0001), presence of basal-like (p < 0.0001) and triple-negative phenotypes (p < 0.0001). Low SMUG1 protein expression was associated with loss of BRCA1 (p < 0.0001), ATM (p < 0.0001) and XRCC1 (p < 0.0001). Low p27 (p < 0.0001), low p21 (p = 0.023), mutant p53 (p = 0.037), low MDM2 (p < 0.0001), low MDM4 (p = 0.004), low Bcl-2 (p = 0.001), low Bax (p = 0.003) and high MIB1 (p < 0.0001) were likely in low SMUG1 tumours. Low SMUG1 protein expression was associated with poor prognosis in univariate (p < 0.001) and multivariate analysis (p < 0.01). In ER+ cohort that received adjuvant endocrine therapy, low SMUG1 protein expression remains associated with poor survival (p < 0.01). In ER- cohort that received adjuvant chemotherapy, low SMUG1 protein expression is associated with improved survival (p = 0.043). Our study suggests that low SMUG1 expression may correlate to adverse clinicopathological features and predict response to adjuvant therapy in breast cancer.


Assuntos
Neoplasias da Mama/genética , Uracila-DNA Glicosidase/deficiência , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Estudos de Coortes , Feminino , Humanos , Gradação de Tumores , Estadiamento de Neoplasias , Fenótipo , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transcrição Gênica , Resultado do Tratamento , Carga Tumoral , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo
15.
J Immunol ; 190(12): 5949-60, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23667108

RESUMO

Activation-induced deaminase converts deoxycytidine to deoxyuridine at the Ig loci. Complementary pathways, initiated by the uracil-DNA glycosylase (UNG) or the mismatch repair factor MSH2/MSH6, must process the deoxyuridine to initiate class-switch recombination (CSR) and somatic hypermutation. UNG deficiency most severely reduces CSR efficiency and only modestly affects the somatic hypermutation spectrum in vitro. This would predict isotype-switching deficiency but normal affinity maturation in Ung(-/-) mice in vivo, but this has not been tested. Moreover, puzzling differences in the amount of circulating Ig between UNG-deficient humans and mice make it unclear to what extent MSH2/MSH6 can complement for UNG in vivo. We find that Ab affinity maturation is indeed unaffected in Ung(-/-) mice, even allowing IgM responses with higher than normal affinity. Ung(-/-) mice display normal to only moderately reduced basal levels of most circulating Ig subclasses and gut-associated IgA, which are elicited in response to chronically available environmental Ag. In contrast, their ability to produce switched Ig in response to immunization or vesicular stomatitis virus infection is strongly impaired. Our results uncover a specific need for UNG in CSR for timely and efficient acute Ab responses in vivo. Furthermore, Ung(-/-) mice provide a novel model for separating isotype switching and affinity maturation during acute (but not chronic) Ab responses, which could be useful for dissecting their relative contribution to some infections. Interestingly, Ung(-/-) mice present with circulating autoantibodies, suggesting that UNG may impinge on tolerance.


Assuntos
Afinidade de Anticorpos/imunologia , Linfócitos B/imunologia , Switching de Imunoglobulina/imunologia , Ativação Linfocitária/imunologia , Uracila-DNA Glicosidase/imunologia , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Uracila-DNA Glicosidase/deficiência
16.
Mol Immunol ; 53(3): 214-7, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22960197

RESUMO

The activation-induced cytidine deaminase (AID) initiates Ig gene hypermutation by converting cytosine to uracil (U) and generating a U:G lesion. Genetic and biochemical studies suggest that the AID-triggered U:G lesions are processed by three mutagenic pathways to induce mutations at both C:G and A:T pairs. First, direct replication of the U:G lesion leads to C to T and G to A transitions. Second, U can be excised by the uracil DNA glycosylase (UNG) and the replication/processing of the resulting abasic site leads to transversions and transitions at C:G pairs. Third, the U:G lesion is recognized by an atypical mismatch repair (MMR) pathway which generates mutations at A:T pairs in a DNA polymerase η (POLH)-dependent manner. To further explore whether these three mutagenic pathways function competitively or independently, we have analyzed Ig gene hypermutation in mice deficient in both UNG and POLH. Compared with WT mice, UNG deficiency caused elevated frequency of C:G mutations, suggesting that UNG-mediated U excision led to error-free as well as error-prone repair. In contrast, UNG deficiency did not affect the frequency and patterns of A:T mutations, suggesting that the MMR did not target U:G lesions normally recognized and processed by UNG. In addition, POLH deficiency did not affect the frequency and patterns of C:G mutations and UNG POLH double deficiency showed an additive effect of single deficiency. Based on these observations and previous results, along with the recent finding that UNG excises AID-triggered U predominantly during G1 phase of the cell cycle, it appears that UNG and MMR targets U:G lesions generated during G1 and S phases of the cell cycle, respectively.


Assuntos
DNA Polimerase Dirigida por DNA/deficiência , DNA Polimerase Dirigida por DNA/genética , Hipermutação Somática de Imunoglobulina , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Sequência de Bases , Citidina Desaminase/metabolismo , Reparo de Erro de Pareamento de DNA/genética , Reparo de Erro de Pareamento de DNA/imunologia , Primers do DNA/genética , Fase G1/genética , Fase G1/imunologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Modelos Imunológicos , Fase S/genética , Fase S/imunologia
17.
Int J Biochem Cell Biol ; 44(9): 1555-68, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22728162

RESUMO

Cells contain low amounts of uracil in DNA which can be the result of dUTP misincorporation during replication or cytosine deamination. Elimination of uracil in the base excision repair pathway yields an abasic site, which is potentially mutagenic unless repaired. The Trypanosoma brucei genome presents a single uracil-DNA glycosylase responsible for removal of uracil from DNA. Here we establish that no excision activity is detected on U:G, U:A pairs or single-strand uracil-containing DNA in uracil-DNA glycosylase null mutant cell extracts, indicating the absence of back-up uracil excision activities. While procyclic forms can survive with moderate amounts of uracil in DNA, an analysis of the mutation rate and spectra in mutant cells revealed a hypermutator phenotype where the predominant events were GC to AT transitions and insertions. Defective elimination of uracil via the base excision repair pathway gives rise to hypersensitivity to antifolates and oxidative stress and an increased number of DNA strand breaks, suggesting the activation of alternative DNA repair pathways. Finally, we show that uracil-DNA glycosylase defective cells exhibit reduced infectivity in vivo demonstrating that efficient uracil elimination is important for survival within the mammalian host.


Assuntos
Antagonistas do Ácido Fólico/farmacologia , Mutação , Fenótipo , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei brucei/enzimologia , Uracila-DNA Glicosidase/deficiência , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Quebras de DNA/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , DNA de Protozoário/genética , Feminino , Sequência Rica em GC , Técnicas de Inativação de Genes , Peróxido de Hidrogênio/farmacologia , Metotrexato/farmacologia , Camundongos , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/patogenicidade , Uracila-DNA Glicosidase/genética
18.
Cell Death Dis ; 3: e252, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22237209

RESUMO

Uracil DNA glycosylase (UDG) specifically removes uracil bases from DNA, and its repair activity determines the sensitivity of the cell to anticancer agents that are capable of introducing uracil into DNA. In the present study, the participation of UDG in the response to pemetrexed-induced incorporation of uracil into DNA was studied using isogenic human tumor cell lines with or without UDG (UDG(+/+)/UDG(-/-)). UDG(-/-) cells were very sensitive to pemetrexed. Cell killing by pemetrexed was associated with genomic uracil accumulation, stalled DNA replication, and catastrophic DNA strand breaks. By contrast, UDG(+/+) cells were >10 times more resistant to pemetrexed due to the rapid removal of uracil from DNA by UDG and subsequent repair of the resultant AP sites (abasic sites) via the base excision repair (BER). The resistance to pemetrexed in UDG(+/+) cells could be reversed by the addition of methoxyamine (MX), which binds to AP sites and interrupts BER pathway. Furthermore, MX-bound AP sites induced cell death was related to their cytotoxic effect of dual inactivation of UDG and topoisomerase IIα, two genes that are highly expressed in lung cancer cells in comparison with normal cells. Thus, targeting BER-based therapy exhibits more selective cytotoxicity on cancer cells through a synthetic lethal mechanism.


Assuntos
Antineoplásicos/farmacologia , Reparo do DNA/efeitos dos fármacos , Expressão Gênica , Glutamatos/farmacologia , Guanina/análogos & derivados , Transdução de Sinais/genética , Uracila-DNA Glicosidase/genética , Animais , Antígenos de Neoplasias/genética , Linhagem Celular Tumoral , DNA/genética , Dano ao DNA , DNA Topoisomerases Tipo II/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Guanina/farmacologia , Humanos , Hidroxilaminas/farmacologia , Camundongos , Camundongos Nus , Pemetrexede , Transdução de Sinais/efeitos dos fármacos , Uracila/metabolismo , Uracila-DNA Glicosidase/antagonistas & inibidores , Uracila-DNA Glicosidase/deficiência , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Proc Natl Acad Sci U S A ; 108(12): 4858-63, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21368204

RESUMO

The human APOBEC3 (A3A-A3H) locus encodes six cytidine deaminases that edit single-stranded DNA, the result being DNA peppered with uridine. Although several cytidine deaminases are clearly restriction factors for retroviruses and hepadnaviruses, it is not known if APOBEC3 enzymes have roles outside of these settings. It is shown here that both human mitochondrial and nuclear DNA are vulnerable to somatic hypermutation by A3 deaminases, with APOBEC3A standing out among them. The degree of editing is much greater in patients lacking the uracil DNA-glycolyase gene, indicating that the observed levels of editing reflect a dynamic composed of A3 editing and DNA catabolism involving uracil DNA-glycolyase. Nonetheless, hyper- and lightly mutated sequences went hand in hand, raising the hypothesis that recurrent low-level mutation by APOBEC3A could catalyze the transition from a healthy to a cancer genome.


Assuntos
Citosina Desaminase/metabolismo , DNA Mitocondrial/metabolismo , Loci Gênicos , Genoma Humano , Mutação , Desaminases APOBEC , Citidina Desaminase , Citosina Desaminase/genética , DNA Mitocondrial/genética , Feminino , Células HeLa , Hepadnaviridae/genética , Hepadnaviridae/metabolismo , Humanos , Masculino , Neoplasias/enzimologia , Neoplasias/genética , Retroviridae/genética , Retroviridae/metabolismo , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo
20.
Exp Neurol ; 228(2): 253-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21281628

RESUMO

Strong epidemiological and experimental evidence links folate deficiency and resultant hyperhomocysteinemia with cognitive decline and neurodegeneration. Here, we tested the hypothesis that uracil misincorporation contributes to mitochondrial pathology in aged brain following folate deprivation. In a 2 × 2 design, 14-month-old mice lacking uracil DNA glycosylase (Ung-/-) versus wild-type controls were subjected to a folate-deficient versus a regular diet for six weeks. Folate-deficient feeding significantly enhanced mtDNA content and overall abundance of the D-1 mtDNA deletion in brain of Ung-/-, but not of wild-type mice. Independent of folate status, the frequency of the D-1 mtDNA deletion in mtDNA was significantly increased in Ung-/- mice. The rate of mitochondrial biogenesis as assessed at six weeks of the experimental diet by mRNA expression levels of transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α and of mitochondrial transcription factor A (Tfam) was not affected by either Ung-/- genotype or short-term folate deficiency. Similarly, citrate synthase (CS) activity in the brain did not differ across experimental groups. By contrast, independent of genotype, lactate dehydrogenase (LDH) activity was significantly reduced in folate-deficient animals. Our results suggest that impaired uracil excision repair causes an increase in mitochondrial mutagenesis in aged brain along with a compensatory increase in mtDNA content in response to low folate status. Folate deficiency may contribute to neurodegeneration via mtDNA damage.


Assuntos
Envelhecimento/genética , Deficiência de Ácido Fólico/genética , Deleção de Genes , Doenças Mitocondriais/genética , Doenças Neurodegenerativas/genética , Uracila-DNA Glicosidase/deficiência , Uracila-DNA Glicosidase/genética , Envelhecimento/metabolismo , Animais , Encefalopatias/complicações , Encefalopatias/enzimologia , Encefalopatias/genética , DNA Mitocondrial/genética , Regulação para Baixo/genética , Deficiência de Ácido Fólico/complicações , Deficiência de Ácido Fólico/diagnóstico , Camundongos , Camundongos Knockout , Doenças Mitocondriais/complicações , Doenças Mitocondriais/enzimologia , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/etiologia , Uracila-DNA Glicosidase/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...