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










Base de dados
Intervalo de ano de publicação
1.
Carcinogenesis ; 43(2): 160-169, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-34919656

RESUMO

The prevalent cancer predisposition Lynch syndrome (LS, OMIM #120435) is caused by an inherited heterozygous defect in any of the four core DNA mismatch repair (MMR) genes MSH2, MSH6, MLH1 or PMS2. MMR repairs errors by the replicative DNA polymerases in all proliferating tissues. Its deficiency, following somatic loss of the wild-type copy, results in a spontaneous mutator phenotype that underlies the rapid development of, predominantly, colorectal cancer (CRC) in LS. Here, we have addressed the hypothesis that aberrant responses of intestinal stem cells to diet-derived mutagens may be causally involved in the restricted cancer tropism of LS. To test this we have generated a panel of isogenic mouse embryonic stem (mES) cells with heterozygous or homozygous disruption of multiple MMR genes and investigated their responses to the common dietary mutagen and carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Our data reveal that PhIP can inactivate the wild-type allele of heterozygous mES cells via the induction of either loss of heterozygosity (LOH) or intragenic mutations. Moreover, while protective DNA damage signaling (DDS) is compromised, PhIP induces more mutations in Msh2, Mlh1, Msh6 or Pms2-deficient mES cells than in wild-type cells. Combined with their spontaneous mutator phenotypes, this results in a compound hypermutator phenotype. Together, these results indicate that dietary mutagens may promote CRC development in LS at multiple levels, providing a rationale for dietary modifications in the management of LS.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose , Animais , Neoplasias Encefálicas , Neoplasias Colorretais , Neoplasias Colorretais Hereditárias sem Polipose/genética , Dano ao DNA , Reparo de Erro de Pareamento de DNA/genética , Proteínas de Ligação a DNA/genética , Dieta/efeitos adversos , Mutação em Linhagem Germinativa , Camundongos , Endonuclease PMS2 de Reparo de Erro de Pareamento/genética , Proteína 1 Homóloga a MutL/genética , Proteína 2 Homóloga a MutS/genética , Mutagênicos/toxicidade , Síndromes Neoplásicas Hereditárias
2.
DNA Repair (Amst) ; 93: 102923, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-33087264

RESUMO

Canonical DNA mismatch repair (MMR) excises base-base mismatches to increase the fidelity of DNA replication. Thus, loss of MMR leads to increased spontaneous mutagenesis. MMR genes also are involved in the suppression of mutagenic, and the induction of protective, responses to various types of DNA damage. In this review we describe these non-canonical roles of MMR at different lesion types. Loss of non-canonical MMR gene functions may have important ramifications for the prevention, development and treatment of colorectal cancer associated with inherited MMR gene defects in Lynch syndrome. This graphical review pays tribute to Samuel H. Wilson. Sam not only made seminal contributions to understanding base excision repair, particularly with respect to structure-function relationships in DNA polymerase ß but also, as Editor of DNA Repair, has maintained a high standard of the journal.


Assuntos
Neoplasias Colorretais/genética , Dano ao DNA , Reparo de Erro de Pareamento de DNA , Neoplasias Colorretais Hereditárias sem Polipose , DNA/metabolismo , Replicação do DNA , Humanos , Mutagênese
5.
Nucleic Acids Res ; 43(17): 8325-39, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26187992

RESUMO

REV1 is a eukaryotic member of the Y-family of DNA polymerases involved in translesion DNA synthesis and genome mutagenesis. Recently, REV1 is also found to function in homologous recombination. However, it remains unclear how REV1 is recruited to the sites where homologous recombination is processed. Here, we report that loss of mammalian REV1 results in a specific defect in replication-associated gene conversion. We found that REV1 is targeted to laser-induced DNA damage stripes in a manner dependent on its ubiquitin-binding motifs, on RAD18, and on monoubiquitinated FANCD2 (FANCD2-mUb) that associates with REV1. Expression of a FANCD2-Ub chimeric protein in RAD18-depleted cells enhances REV1 assembly at laser-damaged sites, suggesting that FANCD2-mUb functions downstream of RAD18 to recruit REV1 to DNA breaks. Consistent with this suggestion we found that REV1 and FANCD2 are epistatic with respect to sensitivity to the double-strand break-inducer camptothecin. REV1 enrichment at DNA damage stripes also partially depends on BRCA1 and BRCA2, components of the FANCD2/BRCA supercomplex. Intriguingly, analogous to FANCD2-mUb and BRCA1/BRCA2, REV1 plays an unexpected role in protecting nascent replication tracts from degradation by stabilizing RAD51 filaments. Collectively these data suggest that REV1 plays multiple roles at stalled replication forks in response to replication stress.


Assuntos
Dano ao DNA , Replicação do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Proteínas Nucleares/fisiologia , Nucleotidiltransferases/fisiologia , Camptotecina/toxicidade , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , DNA Polimerase Dirigida por DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Conversão Gênica , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estresse Fisiológico/genética , Inibidores da Topoisomerase I/toxicidade , Ubiquitina-Proteína Ligases
7.
Nat Commun ; 6: 7199, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26068067

RESUMO

Möbius syndrome (MBS) is a neurological disorder that is characterized by paralysis of the facial nerves and variable other congenital anomalies. The aetiology of this syndrome has been enigmatic since the initial descriptions by von Graefe in 1880 and by Möbius in 1888, and it has been debated for decades whether MBS has a genetic or a non-genetic aetiology. Here, we report de novo mutations affecting two genes, PLXND1 and REV3L in MBS patients. PLXND1 and REV3L represent totally unrelated pathways involved in hindbrain development: neural migration and DNA translesion synthesis, essential for the replication of endogenously damaged DNA, respectively. Interestingly, analysis of Plxnd1 and Rev3l mutant mice shows that disruption of these separate pathways converge at the facial branchiomotor nucleus, affecting either motoneuron migration or proliferation. The finding that PLXND1 and REV3L mutations are responsible for a proportion of MBS patients suggests that de novo mutations in other genes might account for other MBS patients.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Proteínas de Ligação a DNA/genética , DNA Polimerase Dirigida por DNA/genética , Síndrome de Möbius/genética , Mutação , Animais , Dano ao DNA , Exoma , Heterozigoto , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana , Camundongos , Camundongos Mutantes
8.
J Cell Biol ; 209(1): 33-46, 2015 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-25869665

RESUMO

In addition to correcting mispaired nucleotides, DNA mismatch repair (MMR) proteins have been implicated in mutagenic, cell cycle, and apoptotic responses to agents that induce structurally aberrant nucleotide lesions. Here, we investigated the mechanistic basis for these responses by exposing cell lines with single or combined genetic defects in nucleotide excision repair (NER), postreplicative translesion synthesis (TLS), and MMR to low-dose ultraviolet light during S phase. Our data reveal that the MMR heterodimer Msh2/Msh6 mediates the excision of incorrect nucleotides that are incorporated by TLS opposite helix-distorting, noninstructive DNA photolesions. The resulting single-stranded DNA patches induce canonical Rpa-Atr-Chk1-mediated checkpoints and, in the next cell cycle, collapse to double-stranded DNA breaks that trigger apoptosis. In conclusion, a novel MMR-related DNA excision repair pathway controls TLS a posteriori, while initiating cellular responses to environmentally relevant densities of genotoxic lesions. These results may provide a rationale for the colorectal cancer tropism in Lynch syndrome, which is caused by inherited MMR gene defects.


Assuntos
Dano ao DNA , Reparo de Erro de Pareamento de DNA , Animais , Apoptose , Linhagem Celular , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Embrionárias/fisiologia , Epistasia Genética , Humanos , Camundongos da Linhagem 129 , Proteína 2 Homóloga a MutS/fisiologia , Mutagênese
9.
DNA Repair (Amst) ; 29: 56-64, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25655219

RESUMO

Most spontaneous and DNA damage-induced nucleotide substitutions in eukaryotes depend on translesion synthesis polymerases Rev1 and Pol ζ, the latter consisting of the catalytic subunit Rev3 and the accessory protein Rev7. Here we review the regulation, and the biochemical and cellular functions, of Rev1/Pol ζ-dependent translesion synthesis. These are correlated with phenotypes of mouse models with defects in Rev1, Rev3 or Rev7. The data indicate that Rev1/Pol ζ-mediated translesion synthesis is important for adaptive immunity while playing paradoxical roles in oncogenesis. On the other hand, by enabling the replication of endogenously damaged templates, Rev1/Pol ζ -dependent translesion synthesis protects stem cells, thereby preventing features of ageing. In conclusion, Rev1/Pol ζ-dependent translesion synthesis at DNA helix-distorting nucleotide lesions orchestrates pleiotropic responses that determine organismal fitness and disease.


Assuntos
Reparo do DNA/fisiologia , Replicação do DNA/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Instabilidade Genômica , Mutagênese , Animais , DNA/metabolismo , Doença/genética , Proteínas Mad2/metabolismo , Camundongos , Nucleotidiltransferases/metabolismo
10.
Nucleic Acids Res ; 43(1): 282-94, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25505145

RESUMO

Translesion synthesis (TLS) provides a highly conserved mechanism that enables DNA synthesis on a damaged template. TLS is performed by specialized DNA polymerases of which polymerase (Pol) κ is important for the cellular response to DNA damage induced by benzo[a]pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), ultraviolet (UV) light and the alkylating agent methyl methanesulfonate (MMS). As TLS polymerases are intrinsically error-prone, tight regulation of their activity is required. One level of control is provided by ubiquitination of the homotrimeric DNA clamp PCNA at lysine residue 164 (PCNA-Ub). We here show that Polκ can function independently of PCNA modification and that Polη can function as a backup during TLS of MMS-induced lesions. Compared to cell lines deficient for PCNA modification (Pcna(K164R)) or Polκ, double mutant cell lines display hypersensitivity to MMS but not to BPDE or UV-C. Double mutant cells also displayed delayed post-replicative TLS, accumulate higher levels of replication stress and delayed S-phase progression. Furthermore, we show that Polη and Polκ are redundant in the DNA damage bypass of MMS-induced DNA damage. Taken together, we provide evidence for PCNA-Ub-independent activation of Polκ and establish Polη as an important backup polymerase in the absence of Polκ in response to MMS-induced DNA damage.


Assuntos
Dano ao DNA , DNA Polimerase Dirigida por DNA/fisiologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitinação , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Sobrevivência Celular , Células Cultivadas , Quinase 1 do Ponto de Checagem , Replicação do DNA , DNA Polimerase Dirigida por DNA/genética , Metanossulfonato de Metila/toxicidade , Camundongos Knockout , Mutação , Antígeno Nuclear de Célula em Proliferação/genética , Proteínas Quinases/metabolismo , Fase S
11.
Nucleic Acids Res ; 42(17): 11071-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170086

RESUMO

Short-wave ultraviolet light induces both mildly helix-distorting cyclobutane pyrimidine dimers (CPDs) and severely distorting (6-4) pyrimidine pyrimidone photoproducts ((6-4)PPs). The only DNA polymerase (Pol) that is known to replicate efficiently across CPDs is Polη, a member of the Y family of translesion synthesis (TLS) DNA polymerases. Phenotypes of Polη deficiency are transient, suggesting redundancy with other DNA damage tolerance pathways. Here we performed a comprehensive analysis of the temporal requirements of Y-family Pols ι and κ as backups for Polη in (i) bypassing genomic CPD and (6-4)PP lesions in vivo, (ii) suppressing DNA damage signaling, (iii) maintaining cell cycle progression and (iv) promoting cell survival, by using mouse embryonic fibroblast lines with single and combined disruptions in these Pols. The contribution of Polι is restricted to TLS at a subset of the photolesions. Polκ plays a dominant role in rescuing stalled replication forks in Polη-deficient mouse embryonic fibroblasts, both at CPDs and (6-4)PPs. This dampens DNA damage signaling and cell cycle arrest, and results in increased survival. The role of relatively error-prone Pols ι and κ as backups for Polη contributes to the understanding of the mutator phenotype of xeroderma pigmentosum variant, a syndrome caused by Polη defects.


Assuntos
Dano ao DNA , DNA Polimerase Dirigida por DNA/fisiologia , Raios Ultravioleta/efeitos adversos , Animais , Ciclo Celular , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Genoma , Camundongos , Dímeros de Pirimidina/metabolismo , DNA Polimerase iota
12.
Nucleic Acids Res ; 42(7): 4406-13, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24464993

RESUMO

Rev3, the catalytic subunit of DNA polymerase ζ, is essential for translesion synthesis of cytotoxic DNA photolesions, whereas the Rev1 protein plays a noncatalytic role in translesion synthesis. Here, we reveal that mammalian Rev3(-/-) and Rev1(-/-) cell lines additionally display a nucleotide excision repair (NER) defect, specifically during S phase. This defect is correlated with the normal recruitment but protracted persistence at DNA damage sites of factors involved in an early stage of NER, while repair synthesis is affected. Remarkably, the NER defect becomes apparent only at 2 h post-irradiation indicating that Rev3 affects repair synthesis only indirectly, rather than performing an enzymatic role in NER. We provide evidence that the NER defect is caused by scarceness of Replication protein A (Rpa) available to NER, resulting from its sequestration at stalled replication forks. Also the induction of replicative stress using hydroxyurea precludes the accumulation of Rpa at photolesion sites, both in Rev3(-/-) and in wild-type cells. These data support a model in which the limited Rpa pool coordinates replicative stress and NER, resulting in increased cytotoxicity of ultraviolet light when replicative stress exceeds a threshold.


Assuntos
Reparo do DNA , Replicação do DNA , Proteína de Replicação A/metabolismo , Animais , Linhagem Celular , Proliferação de Células , DNA Polimerase Dirigida por DNA/genética , Camundongos , Transcrição Gênica , Raios Ultravioleta/efeitos adversos
13.
Mutat Res ; 751-752: 8-14, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24144844

RESUMO

Ultraviolet radiation is a highly mutagenic agent that damages the DNA by the formation of mutagenic photoproducts at dipyrimidine sites and by oxidative DNA damages via reactive oxygen species (ROS). ROS can also give rise to mutations via oxidation of dNTPs in the nucleotide pool, e.g. 8-oxo-dGTP and 2-OH-dATP and subsequent incorporation during DNA replication. Here we show that expression of human MutT homolog 1 (hMTH1) which sanitizes the nucleotide pool by dephosphorylating oxidized dNTPs, protects against mutagenesis induced by long wave UVA light and by UVB light but not by short wave UVC light. Mutational spectra analyses of UVA-induced mutations at the endogenous Thymidine kinase gene in human lymphoblastoid cells revealed that hMTH1 mainly protects cells from transitions at GC and AT base pairs.


Assuntos
Enzimas Reparadoras do DNA/genética , Mutação/efeitos da radiação , Monoéster Fosfórico Hidrolases/genética , Raios Ultravioleta , Pareamento de Bases/efeitos da radiação , Linhagem Celular , Enzimas Reparadoras do DNA/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Mutagênese/efeitos da radiação , Taxa de Mutação , Nucleotídeos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Timidina Quinase/genética
14.
DNA Repair (Amst) ; 11(6): 550-8, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22521143

RESUMO

Replicative polymerases (Pols) arrest at damaged DNA nucleotides, which induces ubiquitination of the DNA sliding clamp PCNA (PCNA-Ub) and DNA damage signaling. PCNA-Ub is associated with the recruitment or activation of translesion synthesis (TLS) DNA polymerases of the Y family that can bypass the lesions, thereby rescuing replication and preventing replication fork collapse and consequent formation of double-strand DNA breaks. Here, we have used gene-targeted mouse embryonic fibroblasts to perform a comprehensive study of the in vivo roles of PCNA-Ub and of the Y family TLS Pols η, ι, κ, Rev1 and the B family TLS Polζ in TLS and in the suppression of DNA damage signaling and genome instability after exposure to UV light. Our data indicate that TLS Pols ι and κ and the N-terminal BRCT domain of Rev1, that previously was implicated in the regulation of TLS, play minor roles in TLS of DNA photoproducts. PCNA-Ub is critical for an early TLS pathway that replicates both strongly helix-distorting (6-4) pyrimidine-pyrimidone ((6-4)PP) and mildly distorting cyclobutane pyrimidine dimer (CPD) photoproducts. The role of Polη is mainly restricted to early TLS of CPD photoproducts, whereas Rev1 and, in particular, Polζ are essential for the bypass of (6-4)PP photoproducts, both early and late after exposure. Thus, structurally distinct photoproducts at the mammalian genome are bypassed by different TLS Pols in temporally different, PCNA-Ub-dependent and independent fashions.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA , Genoma/efeitos da radiação , Transdução de Sinais/genética , Raios Ultravioleta , Animais , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Linhagem Celular , Proliferação de Células/efeitos da radiação , Replicação do DNA/genética , Replicação do DNA/efeitos da radiação , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Genoma/genética , Histonas/metabolismo , Immunoblotting , Mamíferos/genética , Camundongos , Mutação , Fosforilação , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
15.
Toxicol Sci ; 127(1): 130-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22331492

RESUMO

DNA lesions, induced by genotoxic compounds, block the processive replication fork but can be bypassed by specialized translesion synthesis (TLS) DNA polymerases (Pols). TLS safeguards the completion of replication, albeit at the expense of nucleotide substitution mutations. We studied the in vivo role of individual TLS Pols in cellular responses to benzo[a]pyrene diolepoxide (BPDE), a polycyclic aromatic hydrocarbon, and 4-hydroxynonenal (4-HNE), a product of lipid peroxidation. To this aim, we used mouse embryonic fibroblasts with targeted disruptions in the TLS-associated Pols η, ι, κ, and Rev1 as well as in Rev3, the catalytic subunit of TLS Polζ. After exposure, cellular survival, replication fork progression, DNA damage responses (DDR), and the induction of micronuclei were investigated. The results demonstrate that Rev1, Rev3, and, to a lesser extent, Polη are involved in TLS and the prevention of DDR and of DNA breaks, in response to both agents. Conversely, Polκ and the N-terminal BRCT domain of Rev1 are specifically involved in TLS of BPDE-induced DNA damage. We furthermore describe a novel role of Polι in TLS of 4-HNE-induced DNA damage in vivo. We hypothesize that different sets of TLS polymerases act on structurally different genotoxic DNA lesions in vivo, thereby suppressing genomic instability associated with cancer. Our experimental approach may provide a significant contribution in delineating the molecular bases of the genotoxicity in vivo of different classes of DNA-damaging agents.


Assuntos
7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Aldeídos/toxicidade , Dano ao DNA , DNA Polimerase Dirigida por DNA/metabolismo , Mutagênicos/toxicidade , Animais , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Citocinese , Adutos de DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/genética , Fibroblastos/efeitos dos fármacos , Contaminação de Alimentos , Camundongos , Camundongos Knockout , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Testes para Micronúcleos/métodos
16.
DNA Repair (Amst) ; 10(10): 1051-9, 2011 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-21889916

RESUMO

The generation of high affinity antibodies in B cells critically depends on translesion synthesis (TLS) polymerases that introduce mutations into immunoglobulin genes during somatic hypermutation (SHM). The majority of mutations at A/T base pairs during SHM require ubiquitination of PCNA at lysine 164 (PCNA-Ub), which activates TLS polymerases. By comparing the mutation spectra in B cells of WT, TLS polymerase η (Polη)-deficient, PCNA(K164R)-mutant, and PCNA(K164R);Polη double-mutant mice, we now find that most PCNA-Ub-independent A/T mutagenesis during SHM is mediated by Polη. In addition, upon exposure to various DNA damaging agents, PCNA(K164R) mutant cells display strongly impaired recruitment of TLS polymerases, reduced daughter strand maturation and hypersensitivity. Interestingly, compared to the single mutants, PCNA(K164R);Polη double-mutant cells are dramatically delayed in S phase progression and far more prone to cell death following UV exposure. Taken together, these data support the existence of PCNA ubiquitination-dependent and -independent activation pathways of Polη during SHM and DNA damage tolerance.


Assuntos
Linfócitos B/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Hipermutação Somática de Imunoglobulina/genética , Ubiquitinação , Animais , Linfócitos B/citologia , Ativação Enzimática , Lisina/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese , Mutação , Antígeno Nuclear de Célula em Proliferação/genética , Raios Ultravioleta
17.
DNA Repair (Amst) ; 10(9): 915-25, 2011 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-21752727

RESUMO

Rev1 is a eukaryotic DNA polymerase of the Y family involved in translesion synthesis (TLS), a major damage tolerance pathway that allows DNA replication at damaged templates. Uniquely amongst the Y family polymerases, the N-terminal part of Rev1, dubbed the BRCA1 C-terminal homology (BRCT) region, includes a BRCT domain. While most BRCT domains mediate protein-protein interactions, Rev1 contains a predicted α-helix N-terminal to the BRCT domain and in human Replication Factor C (RFC) such a BRCT region endows the protein with DNA binding capacity. Here, we studied the DNA binding properties of yeast and mouse Rev1. Our results show that the BRCT region of Rev1 specifically binds to a 5' phosphorylated, recessed, primer-template junction. This DNA binding depends on the extra α-helix, N-terminal to the BRCT domain. Surprisingly, a stretch of 20 amino acids N-terminal to the predicted α-helix is also critical for high-affinity DNA binding. In addition to 5' primer-template junction binding, Rev1 efficiently binds to a recessed 3' primer-template junction. These dual DNA binding characteristics are discussed in view of the proposed recruitment of Rev1 by 5' primer-template junctions, downstream of stalled replication forks.


Assuntos
DNA Polimerase Dirigida por DNA/metabolismo , DNA/metabolismo , Nucleotidiltransferases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , DNA/genética , Vetores Genéticos/genética , Camundongos , Dados de Sequência Molecular , Nucleotidiltransferases/genética , Nucleotidiltransferases/isolamento & purificação , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Alinhamento de Sequência
18.
Cell Cycle ; 9(12): 2300-4, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20519944

RESUMO

Genomes encode all RNAs required for life. For this simple reason the genome's stability is a prerequisite for maintaining the fitness of the cell, the organism and its progeny. Paradoxically, any enzymatic transaction at the DNA, including transcription itself, entails the risk of local destabilization of the DNA helix, thereby threatening genomic integrity. Particularly where transcription and replication meet, the genome may be at an increased risk of nucleotide substitution mutations, deletions or rearrangements. This Extra-view sketches our understanding of the different threats that transcription imposes on genome stability. We will focus on recent work highlighting the role of DNA damage in transcription-associated mutagenesis (TAM) in mammalian cells. Furthermore we discuss the possible implications of TAM for human fitness and disease with an emphasis on carcinogenesis. In addition, we propose an updated nomenclature for the mechanistically different forms of TAM.


Assuntos
Replicação do DNA , Instabilidade Genômica/genética , Transcrição Gênica , Animais , DNA/genética , DNA/metabolismo , Dano ao DNA , Reparo do DNA , Humanos
19.
Curr Biol ; 20(2): 170-5, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20045328

RESUMO

Skin cancer is the most ubiquitous cancer type in the Caucasian population, and its incidence is increasing rapidly [1]. Transcribed proliferation-related genes in dermal stem cells are targets for the induction of ultraviolet light (UV)-induced mutations that drive carcinogenesis. We have recently found that transcription of a gene increases its mutability by UV in mammalian stem cells, suggesting a role of transcription in skin carcinogenesis [2]. Here we show that transcription-associated UV-induced nucleotide substitutions are caused by increased deamination of cytosines to uracil within photolesions at the transcribed strand, presumably at sites of stalled transcription complexes. Additionally, via an independent mechanism, transcription of UV-damaged DNA induces the generation of intragenic deletions. We demonstrate that transcription-coupled nucleotide excision repair (TC-NER) provides protection against both classes of transcription-associated mutagenesis. Combined, these results unveil the existence of two mutagenic pathways operating specifically at the transcribed DNA strand of active genes. Moreover, these results uncover a novel role for TC-NER in the suppression of UV-induced genome aberrations and provide a rationale for the efficient induction of apoptosis by stalled transcription complexes.


Assuntos
Citosina/metabolismo , Mutagênese , Transcrição Gênica , Raios Ultravioleta , Desaminação , Hipoxantina Fosforribosiltransferase/genética
20.
Transcription ; 1(2): 95-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21326899

RESUMO

Recent data reveal that gene transcription affects genome stability in mammalian cells. For example, transcription of DNA that is damaged by the most prevalent exogenous genotoxin, UV light, induces nucleotide substitutions and chromosomal instability, collectively called UV-induced transcription-associated mutations (UV-TAM). An important class of UV-TAM consists of nucleotide transitions that are caused by deamination of cytosine-containing photolesions to uracil, presumably occurring at stalled transcription complexes. Transcription-associated deletions and recombinational events after UV exposure may be triggered by collisions of replication forks with stalled transcription complexes. In this Point-of-View we propose that mammalian cells possess two tailored mechanisms to prevent UV-TAM in dermal stem cells. First, the transcription-coupled nucleotide excision repair (TCR) pathway removes lesions at transcribed DNA strands, forming the primary barrier against the mutagenic consequences of transcription at a damaged template. Second, when TCR is absent or when the capacity of TCR is exceeded, persistently stalled transcription complexes induce apoptosis, averting the generation of mutant cells following replication. We hypothesize that TCR and the apoptotic response in conjunction reduce the risk of skin carcinogenesis.


Assuntos
Apoptose , Reparo do DNA , Mutagênese/efeitos da radiação , Transcrição Gênica/genética , Raios Ultravioleta , Animais , Dano ao DNA , Humanos , Modelos Genéticos , Mutagênese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...