Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 18(11): e1010485, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36350851

RESUMO

Telomerase activity is the principal telomere maintenance mechanism in human cancers, however 15% of cancers utilise a recombination-based mechanism referred to as alternative lengthening of telomeres (ALT) that leads to long and heterogenous telomere length distributions. Loss-of-function mutations in the Alpha Thalassemia/Mental Retardation Syndrome X-Linked (ATRX) gene are frequently found in ALT cancers. Here, we demonstrate that the loss of ATRX, coupled with telomere dysfunction during crisis, is sufficient to initiate activation of the ALT pathway and that it confers replicative immortality in human fibroblasts. Additionally, loss of ATRX combined with a telomere-driven crisis in HCT116 epithelial cancer cells led to the initiation of an ALT-like pathway. In these cells, a rapid and precise telomeric elongation and the induction of C-circles was observed; however, this process was transient and the telomeres ultimately continued to erode such that the cells either died or the escape from crisis was associated with telomerase activation. In both of these instances, telomere sequencing revealed that all alleles, irrespective of whether they were elongated, were enriched in variant repeat types, that appeared to be cell-line specific. Thus, our data show that the loss of ATRX combined with telomere dysfunction during crisis induces the ALT pathway in fibroblasts and enables a transient activation of ALT in epithelial cells.


Assuntos
Neoplasias , Telomerase , Talassemia alfa , Humanos , Telomerase/genética , Telomerase/metabolismo , Homeostase do Telômero/genética , Proteína Nuclear Ligada ao X/genética , Talassemia alfa/genética , Telômero/genética , Telômero/metabolismo
2.
Hum Mol Genet ; 29(7): 1083-1095, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31628488

RESUMO

Fanconi anemia (FA) is a chromosome instability syndrome characterized by increased cancer predisposition. Specifically, the FA pathway functions to protect genome stability during DNA replication. The central FA pathway protein, FANCD2, locates to stalled replication forks and recruits homologous recombination (HR) factors such as CtBP interacting protein (CtIP) to promote replication fork restart while suppressing new origin firing. Here, we identify alpha-thalassemia retardation syndrome X-linked (ATRX) as a novel physical and functional interaction partner of FANCD2. ATRX is a chromatin remodeler that forms a complex with Death domain-associated protein 6 (DAXX) to deposit the histone variant H3.3 into specific genomic regions. Intriguingly, ATRX was recently implicated in replication fork recovery; however, the underlying mechanism(s) remained incompletely understood. Our findings demonstrate that ATRX forms a constitutive protein complex with FANCD2 and protects FANCD2 from proteasomal degradation. ATRX and FANCD2 localize to stalled replication forks where they cooperate to recruit CtIP and promote MRE11 exonuclease-dependent fork restart while suppressing the firing of new replication origins. Remarkably, replication restart requires the concerted histone H3 chaperone activities of ATRX/DAXX and FANCD2, demonstrating that coordinated histone H3 variant deposition is a crucial event during the reinitiation of replicative DNA synthesis. Lastly, ATRX also cooperates with FANCD2 to promote the HR-dependent repair of directly induced DNA double-stranded breaks. We propose that ATRX is a novel functional partner of FANCD2 to promote histone deposition-dependent HR mechanisms in S-phase.


Assuntos
Proteínas Correpressoras/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Chaperonas Moleculares/genética , Proteína Nuclear Ligada ao X/genética , Linhagem Celular , Cromatina/genética , Montagem e Desmontagem da Cromatina/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Replicação do DNA/genética , Anemia de Fanconi/patologia , Técnicas de Inativação de Genes/métodos , Histonas/genética , Humanos , Proteína Homóloga a MRE11/genética , Rad51 Recombinase/genética , Reparo de DNA por Recombinação/genética , Transdução de Sinais/genética
3.
Genome Res ; 29(5): 737-749, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30872351

RESUMO

Telomere erosion, dysfunction, and fusion can lead to a state of cellular crisis characterized by large-scale genome instability. We investigated the impact of a telomere-driven crisis on the structural integrity of the genome by undertaking whole-genome sequence analyses of clonal populations of cells that had escaped crisis. Quantification of large-scale structural variants revealed patterns of rearrangement consistent with chromothripsis but formed in the absence of functional nonhomologous end-joining pathways. Rearrangements frequently consisted of short fragments with complex mutational patterns, with a repair topology that deviated from randomness showing preferential repair to local regions or exchange between specific loci. We find evidence of telomere involvement with an enrichment of fold-back inversions demarcating clusters of rearrangements. Our data suggest that chromothriptic rearrangements caused by a telomere crisis arise via a replicative repair process involving template switching.


Assuntos
Cromotripsia , Instabilidade Genômica , Telômero/genética , Inversão Cromossômica/genética , Variações do Número de Cópias de DNA/genética , Reparo do DNA por Junção de Extremidades/genética , Variação Estrutural do Genoma/genética , Células HCT116 , Humanos , Mutação , Neoplasias/genética , Origem de Replicação/genética , Telômero/metabolismo , Telômero/fisiologia , Sequenciamento Completo do Genoma
4.
Mol Cell ; 55(6): 829-842, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25201414

RESUMO

Breakpoint junctions of the chromosomal translocations that occur in human cancers display hallmarks of nonhomologous end-joining (NHEJ). In mouse cells, translocations are suppressed by canonical NHEJ (c-NHEJ) components, which include DNA ligase IV (LIG4), and instead arise from alternative NHEJ (alt-NHEJ). Here we used designer nucleases (ZFNs, TALENs, and CRISPR/Cas9) to introduce DSBs on two chromosomes to study translocation joining mechanisms in human cells. Remarkably, translocations were altered in cells deficient for LIG4 or its interacting protein XRCC4. Translocation junctions had significantly longer deletions and more microhomology, indicative of alt-NHEJ. Thus, unlike mouse cells, translocations in human cells are generated by c-NHEJ. Human cancer translocations induced by paired Cas9 nicks also showed a dependence on c-NHEJ, despite having distinct joining characteristics. These results demonstrate an unexpected and striking species-specific difference for common genomic rearrangements associated with tumorigenesis.


Assuntos
Reparo do DNA por Junção de Extremidades , DNA Ligases/genética , Proteínas de Ligação a DNA/genética , Desoxirribonucleases/fisiologia , Translocação Genética/genética , Animais , Cromossomos Humanos , DNA Ligase Dependente de ATP , Humanos , Camundongos , Deleção de Sequência , Especificidade da Espécie , Células Tumorais Cultivadas
5.
Nucleic Acids Res ; 48(9): 4960-4975, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32232411

RESUMO

G-quadruplexes represent unique roadblocks to DNA replication, which tends to stall at these secondary structures. Although G-quadruplexes can be found throughout the genome, telomeres, due to their G-richness, are particularly predisposed to forming these structures and thus represent difficult-to-replicate regions. Here, we demonstrate that exonuclease 1 (EXO1) plays a key role in the resolution of, and replication through, telomeric G-quadruplexes. When replication forks encounter G-quadruplexes, EXO1 resects the nascent DNA proximal to these structures to facilitate fork progression and faithful replication. In the absence of EXO1, forks accumulate at stabilized G-quadruplexes and ultimately collapse. These collapsed forks are preferentially repaired via error-prone end joining as depletion of EXO1 diverts repair away from error-free homology-dependent repair. Such aberrant repair leads to increased genomic instability, which is exacerbated at chromosome termini in the form of dysfunction and telomere loss.


Assuntos
Enzimas Reparadoras do DNA/fisiologia , Replicação do DNA , Exodesoxirribonucleases/fisiologia , Quadruplex G , Telômero/química , Aminoquinolinas/farmacologia , Linhagem Celular , Reparo do DNA por Junção de Extremidades , Reparo do DNA , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Quadruplex G/efeitos dos fármacos , Técnicas de Inativação de Genes , Células HeLa , Humanos , Neoplasias/metabolismo , Neoplasias/mortalidade , Ácidos Picolínicos/farmacologia , Prognóstico
6.
Nucleic Acids Res ; 47(17): 8927-8940, 2019 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-31378812

RESUMO

The maintenance of telomere length is critical to longevity and survival. Specifically, the failure to properly replicate, resect, and/or form appropriate telomeric structures drives telomere shortening and, in turn, genomic instability. The endonuclease CtIP is a DNA repair protein that is well-known to promote genome stability through the resection of endogenous DNA double-stranded breaks. Here, we describe a novel role for CtIP. We show that in the absence of CtIP, human telomeres shorten rapidly to non-viable lengths. This telomere dysfunction results in an accumulation of fusions, breaks, and frank telomere loss. Additionally, CtIP suppresses the generation of circular, extrachromosomal telomeric DNA. These latter structures appear to arise from arrested DNA replication forks that accumulate in the absence of CtIP. Hence, CtIP is required for faithful replication through telomeres via its roles at stalled replication tracts. Our findings demonstrate a new role for CtIP as a protector of human telomere integrity.


Assuntos
Proteínas de Transporte/metabolismo , Quebras de DNA de Cadeia Dupla , Instabilidade Genômica , Proteínas Nucleares/metabolismo , Encurtamento do Telômero/genética , Telômero/metabolismo , Reparo do DNA por Junção de Extremidades , Replicação do DNA , DNA Circular/metabolismo , Endodesoxirribonucleases , Humanos , Telômero/enzimologia , Proteínas de Ligação a Telômeros/metabolismo
7.
Nucleic Acids Res ; 47(18): 9467-9479, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31396623

RESUMO

The DNA damage response (DDR) encompasses the cellular response to DNA double-stranded breaks (DSBs), and includes recognition of the DSB, recruitment of numerous factors to the DNA damage site, initiation of signaling cascades, chromatin remodeling, cell-cycle checkpoint activation, and repair of the DSB. Key drivers of the DDR are multiple members of the phosphatidylinositol 3-kinase-related kinase family, including ataxia telangiectasia mutated (ATM), ataxia telangiectasia and Rad3-related (ATR), and the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). ATM and ATR modulate multiple portions of the DDR, but DNA-PKcs is believed to primarily function in the DSB repair pathway, non-homologous end joining. Utilizing a human cell line in which the kinase domain of DNA-PKcs is inactivated, we show here that DNA-PKcs kinase activity is required for the cellular response to DSBs immediately after their induction. Specifically, DNA-PKcs kinase activity initiates phosphorylation of the chromatin factors H2AX and KAP1 following ionizing radiation exposure and drives local chromatin decondensation near the DSB site. Furthermore, loss of DNA-PKcs kinase activity results in a marked decrease in the recruitment of numerous members of the DDR machinery to DSBs. Collectively, these results provide clear evidence that DNA-PKcs activity is pivotal for the initiation of the DDR.


Assuntos
Cromatina/genética , Dano ao DNA/genética , Reparo do DNA/genética , DNA/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Pontos de Checagem do Ciclo Celular/genética , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Dano ao DNA/efeitos da radiação , Proteína Quinase Ativada por DNA/genética , Humanos , Proteínas Nucleares/genética , Fosforilação/efeitos da radiação , Radiação Ionizante , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação
8.
Nucleic Acids Res ; 47(5): 2402-2424, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30590694

RESUMO

Fusion of critically short or damaged telomeres is associated with the genomic rearrangements that support malignant transformation. We have demonstrated the fundamental contribution of DNA ligase 4-dependent classical non-homologous end-joining to long-range inter-chromosomal telomere fusions. In contrast, localized genomic recombinations initiated by sister chromatid fusion are predominantly mediated by alternative non-homologous end-joining activity that may employ either DNA ligase 3 or DNA ligase 1. In this study, we sought to discriminate the relative involvement of these ligases in sister chromatid telomere fusion through a precise genetic dissociation of functional activity. We have resolved an essential and non-redundant role for DNA ligase 1 in the fusion of sister chromatids bearing targeted double strand DNA breaks that is entirely uncoupled from its requisite engagement in DNA replication. Importantly, this fusogenic repair occurs in cells fully proficient for non-homologous end-joining and is not compensated by DNA ligases 3 or 4. The dual functions of DNA ligase 1 in replication and non-homologous end-joining uniquely position and capacitate this ligase for DNA repair at stalled replication forks, facilitating mitotic progression.


Assuntos
Cromátides/genética , Reparo do DNA por Junção de Extremidades/genética , DNA Ligase Dependente de ATP/genética , Mitose/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Replicação do DNA/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Células HCT116 , Humanos , Proteínas de Ligação a Poli-ADP-Ribose/genética , Troca de Cromátide Irmã/genética , Telômero/genética
9.
Genome Res ; 27(7): 1099-1111, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28356322

RESUMO

The use of programmable meganucleases is transforming genome editing and functional genomics. CRISPR/Cas9 was developed such that targeted genomic lesions could be introduced in vivo with unprecedented ease. In the presence of homology donors, these lesions facilitate high-efficiency precise genome editing (PGE) via homology-directed repair (HDR) pathways. However, the identity and hierarchy of the HDR (sub)pathways leading to the formation of PGE products remain elusive. Here, we established a green to blue fluorescent protein conversion system to systematically characterize oligodeoxynucleotide (ODN)-mediated PGE using Cas9 and its nickase variants in human cells. We demonstrate that, unlike double-stranded DNA (dsDNA) donors with central heterologies, ODNs generated short conversion tracts with Gaussian-like distributions. Interestingly, single-nick-induced PGE using ODN donors produced conversion tracts biased either mostly uni- or bidirectional depending on the relative strandedness of the ODNs and the nick. Moreover, the ODNs were physically incorporated into the genome only in the bidirectional, but not in the unidirectional, conversion pathway. In the presence of double-stranded genomic lesions, the unidirectional conversion pathway was preferentially utilized even though the knock-in mutation could theoretically have been converted by both pathways. Collectively, our results suggest that ODN-mediated PGE utilizes synthesis-dependent strand annealing and single-stranded DNA incorporation pathways. Both of these pathways generate short conversion tracts with Gaussian-like distributions. Although synthesis-dependent strand annealing is preferentially utilized, our work unequivocally establishes the existence of a single-stranded DNA incorporation pathway in human cells. This work extends the paradigms of HDR-mediated gene conversion and establishes guidelines for PGE in human cells.


Assuntos
Sistemas CRISPR-Cas , DNA , Edição de Genes/métodos , Oligodesoxirribonucleotídeos/química , Linhagem Celular , DNA/genética , DNA/metabolismo , Humanos , Oligodesoxirribonucleotídeos/genética
10.
Nucleic Acids Res ; 46(17): 8926-8939, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30113698

RESUMO

The Artemis nuclease and tyrosyl-DNA phosphodiesterase (TDP1) are each capable of resolving protruding 3'-phosphoglycolate (PG) termini of DNA double-strand breaks (DSBs). Consequently, both a knockout of Artemis and a knockout/knockdown of TDP1 rendered cells sensitive to the radiomimetic agent neocarzinostatin (NCS), which induces 3'-PG-terminated DSBs. Unexpectedly, however, a knockdown or knockout of TDP1 in Artemis-null cells did not confer any greater sensitivity than either deficiency alone, indicating a strict epistasis between TDP1 and Artemis. Moreover, a deficiency in Artemis, but not TDP1, resulted in a fraction of unrepaired DSBs, which were assessed as 53BP1 foci. Conversely, a deficiency in TDP1, but not Artemis, resulted in a dramatic increase in dicentric chromosomes following NCS treatment. An inhibitor of DNA-dependent protein kinase, a key regulator of the classical nonhomologous end joining (C-NHEJ) pathway sensitized cells to NCS, but eliminated the sensitizing effects of both TDP1 and Artemis deficiencies. These results suggest that TDP1 and Artemis perform different functions in the repair of terminally blocked DSBs by the C-NHEJ pathway, and that whereas an Artemis deficiency prevents end joining of some DSBs, a TDP1 deficiency tends to promote DSB mis-joining.


Assuntos
Reparo do DNA por Junção de Extremidades , DNA/genética , Endonucleases/genética , Epistasia Genética , Proteínas Nucleares/genética , Diester Fosfórico Hidrolases/genética , Sobrevivência Celular/efeitos dos fármacos , Citotoxinas/farmacologia , DNA/química , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA , Endonucleases/antagonistas & inibidores , Endonucleases/deficiência , Células HCT116 , Células HEK293 , Humanos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/deficiência , Inibidores da Síntese de Ácido Nucleico/farmacologia , Diester Fosfórico Hidrolases/deficiência , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Zinostatina/farmacologia
11.
EMBO J ; 34(10): 1399-416, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25820262

RESUMO

Mutations of CSB account for the majority of Cockayne syndrome (CS), a devastating hereditary disorder characterized by physical impairment, neurological degeneration and segmental premature aging. Here we report the generation of a human CSB-knockout cell line. We find that CSB facilitates HR and represses NHEJ. Loss of CSB or a CS-associated CSB mutation abrogating its ATPase activity impairs the recruitment of BRCA1, RPA and Rad51 proteins to damaged chromatin but promotes the formation of 53BP1-Rif1 damage foci in S and G2 cells. Depletion of 53BP1 rescues the formation of BRCA1 damage foci in CSB-knockout cells. In addition, knockout of CSB impairs the ATM- and Chk2-mediated DNA damage responses, promoting a premature entry into mitosis. Furthermore, we show that CSB accumulates at sites of DNA double-strand breaks (DSBs) in a transcription-dependent manner. The kinetics of DSB-induced chromatin association of CSB is distinct from that of its UV-induced chromatin association. These results reveal novel, important functions of CSB in regulating the DNA DSB repair pathway choice as well as G2/M checkpoint activation.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Linhagem Celular , Células Cultivadas , Dano ao DNA/genética , Humanos , Cinética , Modelos Biológicos , Proteínas de Ligação a Poli-ADP-Ribose , Ubiquitina-Proteína Ligases/genética
12.
Genome Res ; 26(5): 588-600, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26941250

RESUMO

Telomeres shorten with each cell division and can ultimately become substrates for nonhomologous end-joining repair, leading to large-scale genomic rearrangements of the kind frequently observed in human cancers. We have characterized more than 1400 telomere fusion events at the single-molecule level, using a combination of high-throughput sequence analysis together with experimentally induced telomeric double-stranded DNA breaks. We show that a single chromosomal dysfunctional telomere can fuse with diverse nontelomeric genomic loci, even in the presence of an otherwise stable genome, and that fusion predominates in coding regions. Fusion frequency was markedly increased in the absence of TP53 checkpoint control and significantly modulated by the cellular capacity for classical, versus alternative, nonhomologous end joining (NHEJ). We observed a striking reduction in inter-chromosomal fusion events in cells lacking DNA ligase 4, in contrast to a remarkably consistent profile of intra-chromosomal fusion in the context of multiple genetic knockouts, including DNA ligase 3 and 4 double-knockouts. We reveal distinct mutational signatures associated with classical NHEJ-mediated inter-chromosomal, as opposed to alternative NHEJ-mediated intra-chromosomal, telomere fusions and evidence for an unanticipated sufficiency of DNA ligase 1 for these intra-chromosomal events. Our findings have implications for mechanisms driving cancer genome evolution.


Assuntos
Cromátides , Cromossomos Humanos , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP , Neoplasias , Telômero , Linhagem Celular Tumoral , Cromátides/genética , Cromátides/metabolismo , Cromossomos Humanos/genética , Cromossomos Humanos/metabolismo , Deleção de Genes , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Telômero/genética , Telômero/metabolismo , Proteína Supressora de Tumor p53
13.
Nucleic Acids Res ; 45(20): 11837-11857, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29059323

RESUMO

Fanconi anemia (FA) is an inherited cancer predisposition syndrome characterized by cellular hypersensitivity to DNA interstrand crosslinks (ICLs). To repair these lesions, the FA proteins act in a linear hierarchy: following ICL detection on chromatin, the FA core complex monoubiquitinates and recruits the central FANCI and FANCD2 proteins that subsequently coordinate ICL removal and repair of the ensuing DNA double-stranded break by homology-dependent repair (HDR). FANCD2 also functions during the replication stress response by mediating the restart of temporarily stalled replication forks thereby suppressing the firing of new replication origins. To address if FANCI is also involved in these FANCD2-dependent mechanisms, we generated isogenic FANCI-, FANCD2- and FANCI:FANCD2 double-null cells. We show that FANCI and FANCD2 are partially independent regarding their protein stability, nuclear localization and chromatin recruitment and contribute independently to cellular proliferation. Simultaneously, FANCD2-but not FANCI-plays a major role in HDR-mediated replication restart and in suppressing new origin firing. Consistent with this observation, deficiencies in HDR-mediated DNA DSB repair can be overcome by stabilizing RAD51 filament formation in cells lacking functional FANCD2. We propose that FANCI and FANCD2 have partially non-overlapping and possibly even opposing roles during the replication stress response.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Replicação do DNA , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Sequência de Bases , Ciclo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células/genética , Cromatina/genética , Cromatina/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Células HCT116 , Humanos , Immunoblotting , Mutação , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Homologia de Sequência do Ácido Nucleico
14.
Adv Exp Med Biol ; 1044: 89-112, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29956293

RESUMO

Chromosomal translocations are now well understood to not only constitute signature molecular markers for certain human cancers but often also to be causative in the genesis of that tumor. Despite the obvious importance of such events, the molecular mechanism of chromosomal translocations in human cells remains poorly understood. Part of the explanation for this dearth of knowledge is due to the complexity of the reaction and the need to archaeologically work backwards from the final product (a translocation) to the original unrearranged chromosomes to infer mechanism. Although not definitive, these studies have indicated that the aberrant usage of endogenous DNA repair pathways likely lies at the heart of the problem. An equally obfuscating aspect of this field, however, has also originated from the unfortunate species-specific differences that appear to exist in the relevant model systems that have been utilized to investigate this process. Specifically, yeast and murine systems (which are often used by basic science investigators) rely on different DNA repair pathways to promote chromosomal translocations than human somatic cells. In this chapter, we will review some of the basic concepts of chromosomal translocations and the DNA repair systems thought to be responsible for their genesis with an emphasis on underscoring the differences between other species and human cells. In addition, we will focus on a specific subset of translocations that involve the very end of a chromosome (a telomere). A better understanding of the relationship between DNA repair pathways and chromosomal translocations is guaranteed to lead to improved therapeutic treatments for cancer.


Assuntos
DNA Ligases/fisiologia , Telômero , Translocação Genética , Animais , Dano ao DNA , Reparo do DNA , Humanos , Camundongos
15.
PLoS Genet ; 11(10): e1005563, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26430909

RESUMO

The Fanconi anemia (FA)-BRCA pathway mediates repair of DNA interstrand crosslinks. The FA core complex, a multi-subunit ubiquitin ligase, participates in the detection of DNA lesions and monoubiquitinates two downstream FA proteins, FANCD2 and FANCI (or the ID complex). However, the regulation of the FA core complex itself is poorly understood. Here we show that the FA core complex proteins are recruited to sites of DNA damage and form nuclear foci in S and G2 phases of the cell cycle. ATR kinase activity, an intact FA core complex and FANCM-FAAP24 were crucial for this recruitment. Surprisingly, FANCI, but not its partner FANCD2, was needed for efficient FA core complex foci formation. Monoubiquitination or ATR-dependent phosphorylation of FANCI were not required for the FA core complex recruitment, but FANCI deubiquitination by USP1 was. Additionally, BRCA1 was required for efficient FA core complex foci formation. These findings indicate that FANCI functions upstream of FA core complex recruitment independently of FANCD2, and alter the current view of the FA-BRCA pathway.


Assuntos
Dano ao DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Anemia de Fanconi/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Medula Óssea/patologia , Cromatina/genética , Anemia de Fanconi/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Células HCT116 , Humanos , Fosforilação , RNA Interferente Pequeno , Transdução de Sinais , Ubiquitinação/genética
16.
PLoS Genet ; 10(4): e1004251, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699519

RESUMO

Gene targeting in human somatic cells is of importance because it can be used to either delineate the loss-of-function phenotype of a gene or correct a mutated gene back to wild-type. Both of these outcomes require a form of DNA double-strand break (DSB) repair known as homologous recombination (HR). The mechanism of HR leading to gene targeting, however, is not well understood in human cells. Here, we demonstrate that a two-end, ends-out HR intermediate is valid for human gene targeting. Furthermore, the resolution step of this intermediate occurs via the classic DSB repair model of HR while synthesis-dependent strand annealing and Holliday Junction dissolution are, at best, minor pathways. Moreover, and in contrast to other systems, the positions of Holliday Junction resolution are evenly distributed along the homology arms of the targeting vector. Most unexpectedly, we demonstrate that when a meganuclease is used to introduce a chromosomal DSB to augment gene targeting, the mechanism of gene targeting is inverted to an ends-in process. Finally, we demonstrate that the anti-recombination activity of mismatch repair is a significant impediment to gene targeting. These observations significantly advance our understanding of HR and gene targeting in human cells.


Assuntos
Reparo do DNA/genética , DNA/genética , Recombinação Genética/genética , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Cruciforme/genética , Marcação de Genes/métodos , Vetores Genéticos/genética , Células HCT116 , Humanos
17.
Hum Mol Genet ; 23(14): 3695-705, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24556218

RESUMO

Fanconi anemia (FA) is a chromosome instability syndrome characterized by increased cancer predisposition. Within the FA pathway, an upstream FA core complex mediates monoubiquitination and recruitment of the central FANCD2 protein to sites of stalled replication forks. Once recruited, FANCD2 fulfills a dual role towards replication fork recovery: (i) it cooperates with BRCA2 and RAD51 to protect forks from nucleolytic degradation and (ii) it recruits the BLM helicase to promote replication fork restart while suppressing new origin firing. Intriguingly, FANCD2 and its interaction partners are also involved in homologous recombination (HR) repair of DNA double-strand breaks, hinting that FANCD2 utilizes HR proteins to mediate replication fork recovery. One such candidate is CtIP (CtBP-interacting protein), a key HR repair factor that functions in complex with BRCA1 and MRE11, but has not been investigated as putative player in the replication stress response. Here, we identify CtIP as a novel interaction partner of FANCD2. CtIP binds and stabilizes FANCD2 in a DNA damage- and FA core complex-independent manner, suggesting that FANCD2 monoubiquitination is dispensable for its interaction with CtIP. Following cellular treatment with a replication inhibitor, aphidicolin, FANCD2 recruits CtIP to transiently stalled, as well as collapsed, replication forks on chromatin. At stalled forks, CtIP cooperates with FANCD2 to promote fork restart and the suppression of new origin firing. Both functions are dependent on BRCA1 that controls the step-wise recruitment of MRE11, FANCD2 and finally CtIP to stalled replication forks, followed by their concerted actions to promote fork recovery.


Assuntos
Proteína BRCA1/metabolismo , Proteínas de Transporte/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Proteínas Nucleares/metabolismo , Afidicolina/farmacologia , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Endodesoxirribonucleases , Anemia de Fanconi/metabolismo , Regulação da Expressão Gênica , Humanos , Proteína Homóloga a MRE11 , Ubiquitinação
18.
Nucleic Acids Res ; 41(3): 1734-49, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23275564

RESUMO

Classic non-homologous end joining (C-NHEJ) is the predominant DNA double-strand break repair pathway in humans. Although seven genes Ku70, Ku86, DNA-PK(cs), Artemis, DNA Ligase IV (LIGIV), X-ray cross-complementing group 4 and XRCC4-like factor are required for C-NHEJ, several of them also have ancillary functions. For example, Ku70:Ku86 possesses an essential telomere maintenance activity. In contrast, LIGIV is believed to function exclusively in C-NHEJ. Moreover, a viable LIGIV-null human B-cell line and LIGIV-reduced patient cell lines have been described. Together, these observations suggest that LIGIV (and hence C-NHEJ), albeit important, is nonetheless dispensable, whereas Ku70:Ku86 and telomere maintenance are essential. To confirm this hypothesis, we inactivated LIGIV in the epithelial human cell line, HCT116. The resulting LIGIV-null cell line was viable, verifying that the gene and C-NHEJ are not essential. However, functional inactivation of RAD54B, a key homologous recombination factor, in the LIGIV-null background yielded no viable clones, suggesting that the combined absence of RAD54B/homologous recombination and C-NHEJ is synthetically lethal. Finally, we demonstrate that LIGIV is differentially required for certain chromosome fusion events induced by telomere dysfunction-used for those owing to the overexpression of a dominant negative version of telomere recognition factor 2, but not used for those owing to absence of Ku70:Ku86.


Assuntos
Aberrações Cromossômicas , Reparo do DNA por Junção de Extremidades , DNA Helicases/genética , DNA Ligases/fisiologia , Proteínas Nucleares/genética , Telômero/fisiologia , Antígenos Nucleares/genética , Linhagem Celular Tumoral , Proliferação de Células , Cromátides , Dano ao DNA , DNA Ligase Dependente de ATP , DNA Ligases/genética , DNA Ligases/metabolismo , Proteínas de Ligação a DNA/genética , Marcação de Genes , Instabilidade Genômica , Humanos , Autoantígeno Ku , Mutação , Recombinação Genética , Reparo de DNA por Recombinação , Homeostase do Telômero , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
19.
Nucleic Acids Res ; 41(4): 2296-312, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23295675

RESUMO

The E3 ubiquitin ligase Rad18 mediates tolerance of replication fork-stalling bulky DNA lesions, but whether Rad18 mediates tolerance of bulky DNA lesions acquired outside S-phase is unclear. Using synchronized cultures of primary human cells, we defined cell cycle stage-specific contributions of Rad18 to genome maintenance in response to ultraviolet C (UVC) and H(2)O(2)-induced DNA damage. UVC and H(2)O(2) treatments both induced Rad18-mediated proliferating cell nuclear antigen mono-ubiquitination during G(0), G(1) and S-phase. Rad18 was important for repressing H(2)O(2)-induced (but not ultraviolet-induced) double strand break (DSB) accumulation and ATM S1981 phosphorylation only during G(1), indicating a specific role for Rad18 in processing of oxidative DNA lesions outside S-phase. However, H(2)O(2)-induced DSB formation in Rad18-depleted G1 cells was not associated with increased genotoxin sensitivity, indicating that back-up DSB repair mechanisms compensate for Rad18 deficiency. Indeed, in DNA LigIV-deficient cells Rad18-depletion conferred H(2)O(2)-sensitivity, demonstrating functional redundancy between Rad18 and non-homologous end joining for tolerance of oxidative DNA damage acquired during G(1). In contrast with G(1)-synchronized cultures, S-phase cells were H(2)O(2)-sensitive following Rad18-depletion. We conclude that although Rad18 pathway activation by oxidative lesions is not restricted to S-phase, Rad18-mediated trans-lesion synthesis by Polη is dispensable for damage-tolerance in G(1) (because of back-up non-homologous end joining-mediated DSB repair), yet Rad18 is necessary for damage tolerance during S-phase.


Assuntos
Ciclo Celular/genética , Reparo do DNA , Proteínas de Ligação a DNA/fisiologia , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Dano ao DNA , DNA Polimerase Dirigida por DNA/metabolismo , Fase G1/genética , Humanos , Peróxido de Hidrogênio/toxicidade , Oxirredução , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteína de Replicação A/metabolismo , Fase S/genética , Ubiquitina-Proteína Ligases , Ubiquitinação
20.
PLoS Pathog ; 8(8): e1002862, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912580

RESUMO

Production of concatemeric DNA is an essential step during HSV infection, as the packaging machinery must recognize longer-than-unit-length concatemers; however, the mechanism by which they are formed is poorly understood. Although it has been proposed that the viral genome circularizes and rolling circle replication leads to the formation of concatemers, several lines of evidence suggest that HSV DNA replication involves recombination-dependent replication reminiscent of bacteriophages λ and T4. Similar to λ, HSV-1 encodes a 5'-to-3' exonuclease (UL12) and a single strand annealing protein [SSAP (ICP8)] that interact with each other and can perform strand exchange in vitro. By analogy with λ phage, HSV may utilize viral and/or cellular recombination proteins during DNA replication. At least four double strand break repair pathways are present in eukaryotic cells, and HSV-1 is known to manipulate several components of these pathways. Chromosomally integrated reporter assays were used to measure the repair of double strand breaks in HSV-infected cells. Single strand annealing (SSA) was increased in HSV-infected cells, while homologous recombination (HR), non-homologous end joining (NHEJ) and alternative non-homologous end joining (A-NHEJ) were decreased. The increase in SSA was abolished when cells were infected with a viral mutant lacking UL12. Moreover, expression of UL12 alone caused an increase in SSA, which was completely eliminated when a UL12 mutant lacking exonuclease activity was expressed. UL12-mediated stimulation of SSA was decreased in cells lacking the cellular SSAP, Rad52, and could be restored by coexpressing the viral SSAP, ICP8, indicating that an SSAP is also required. These results demonstrate that UL12 can specifically stimulate SSA and that either ICP8 or Rad52 can function as an SSAP. We suggest that SSA is the homology-mediated repair pathway utilized during HSV infection.


Assuntos
Replicação do DNA , DNA de Cadeia Simples/metabolismo , DNA Viral/biossíntese , Desoxirribonucleases/metabolismo , Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Recombinação Homóloga , Proteínas Virais/metabolismo , DNA de Cadeia Simples/genética , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desoxirribonucleases/genética , Células HEK293 , Herpes Simples/genética , Humanos , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa