Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 5044, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890315

RESUMO

Homology-dependent targeted DNA integration, generally referred to as gene targeting, provides a powerful tool for precise genome modification; however, its fundamental mechanisms remain poorly understood in human cells. Here we reveal a noncanonical gene targeting mechanism that does not rely on the homologous recombination (HR) protein Rad51. This mechanism is suppressed by Rad52 inhibition, suggesting the involvement of single-strand annealing (SSA). The SSA-mediated gene targeting becomes prominent when DSB repair by HR or end-joining pathways is defective and does not require isogenic DNA, permitting 5% sequence divergence. Intriguingly, loss of Msh2, loss of BLM, and induction of a target-site DNA break all significantly and synergistically enhance SSA-mediated targeted integration. Most notably, SSA-mediated integration is cell cycle-independent, occurring in the G1 phase as well. Our findings provide unequivocal evidence for Rad51-independent targeted integration and unveil multiple mechanisms to regulate SSA-mediated targeted as well as random integration.


Assuntos
Ciclo Celular , Marcação de Genes , Proteína 2 Homóloga a MutS , Rad51 Recombinase , Proteína Rad52 de Recombinação e Reparo de DNA , Humanos , Rad51 Recombinase/metabolismo , Rad51 Recombinase/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Ciclo Celular/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteína 2 Homóloga a MutS/genética , RecQ Helicases/metabolismo , RecQ Helicases/genética , Recombinação Homóloga , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Reparo do DNA por Junção de Extremidades , Fase G1/genética
2.
FEBS Open Bio ; 14(1): 37-50, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37953493

RESUMO

Male and female reproductive tracts develop from anterior intermediate mesoderm with similar differentiation processes. The anterior intermediate mesoderm develops into the mesonephros, and the Wolffian duct initiates by epithelialization in the mesonephros. The Müllerian duct invaginates from the coelomic epithelium of the cranial mesonephros for ductal formation and is then regionalized into proximal to caudal female reproductive tracts. In this study, we focused on the epithelialization of the Wolffian duct, initiation of the Müllerian duct, and the regionalization step of the Müllerian ducts as a continuous process. By using intermediate mesodermal cells from mouse pluripotent stem cells, we identified that inhibition of SMAD2/3 signaling might be involved in the differentiation into mesenchymal cells, after which mesonephric cells might be then epithelialized during differentiation of the Wolffian duct. Aggregation of coelomic epithelial cells might be related to initiation of the Müllerian duct. Transcriptomic analysis predicted that consensus sequences of SMAD3/4 were enriched among highly expressed genes in the proximal Müllerian duct. SMAD2/3 signaling to regulate differentiation of the Wolffian duct was continuously activated in the proximal Müllerian duct and was involved in proximal and oviductal regionalization. Therefore, SMAD2/3 signaling may be finely tuned to regulate differentiation from initiation to regionalization steps.


Assuntos
Ductos Paramesonéfricos , Ductos Mesonéfricos , Camundongos , Animais , Masculino , Feminino , Ductos Mesonéfricos/fisiologia , Ductos Paramesonéfricos/fisiologia , Diferenciação Celular , Células Epiteliais , Transdução de Sinais
3.
FEBS J ; 290(22): 5313-5321, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37530740

RESUMO

Arsenic is a carcinogen that can cause skin, lung, and bladder cancer. While DNA double-strand breaks (DSBs) have been implicated in arsenic-induced carcinogenesis, the exact mechanism remains unclear. In this study, we performed genetic analysis to examine the impact of arsenic trioxide (As2 O3 ) on four different DSB repair pathways using the human pre-B cell line Nalm-6. Random integration analysis showed that As2 O3 does not negatively affect non-homologous end joining or polymerase theta-mediated end joining. In contrast, chromosomal DSB repair analysis revealed that As2 O3 decreases the efficiency of homologous recombination (HR) and, less prominently, single-strand annealing. Consistent with this finding, As2 O3 decreased gene-targeting efficiency, owing to a significant reduction in the frequency of HR-mediated targeted integration. To further verify the inhibitory effect of arsenic on HR, we examined cellular sensitivity to olaparib and camptothecin, which induce one-ended DSBs requiring HR for precise repair. Intriguingly, we found that As2 O3 significantly enhances sensitivity to those anticancer agents in HR-proficient cells. Our results suggest that arsenic-induced genomic instability is attributed to HR suppression, providing valuable insights into arsenic-associated carcinogenesis and therapeutic options.


Assuntos
Arsênio , Quebras de DNA de Cadeia Dupla , Humanos , Reparo do DNA , Recombinação Homóloga , Reparo do DNA por Junção de Extremidades , DNA , Carcinogênese
4.
Sci Rep ; 8(1): 10344, 2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985428

RESUMO

DNA topoisomerase II (Topo II) is crucial for resolving topological problems of DNA and plays important roles in various cellular processes, such as replication, transcription, and chromosome segregation. Although DNA topology problems may also occur during DNA repair, the possible involvement of Topo II in this process remains to be fully investigated. Here, we show the dynamic behavior of human Topo IIß in response to DNA double-strand breaks (DSBs), which is the most harmful form of DNA damage. Live cell imaging coupled with site-directed DSB induction by laser microirradiation demonstrated rapid recruitment of EGFP-tagged Topo IIß to the DSB site. Detergent extraction followed by immunofluorescence showed the tight association of endogenous Topo IIß with DSB sites. Photobleaching analysis revealed that Topo IIß is highly mobile in the nucleus. The Topo II catalytic inhibitors ICRF-187 and ICRF-193 reduced the Topo IIß mobility and thereby prevented Topo IIß recruitment to DSBs. Furthermore, Topo IIß knockout cells exhibited increased sensitivity to bleomycin and decreased DSB repair mediated by homologous recombination (HR), implicating the role of Topo IIß in HR-mediated DSB repair. Taken together, these results highlight a novel aspect of Topo IIß functions in the cellular response to DSBs.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Topoisomerases Tipo II/metabolismo , Bleomicina/toxicidade , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/deficiência , DNA Topoisomerases Tipo II/genética , Dexrazoxano/farmacologia , Células HeLa , Recombinação Homóloga/efeitos dos fármacos , Humanos , Lasers , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
5.
Nat Commun ; 8: 16112, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28695890

RESUMO

Homologous recombination-mediated gene targeting has greatly contributed to genetic analysis in a wide range of species, but is highly inefficient in human cells because of overwhelmingly frequent random integration events, whose molecular mechanism remains elusive. Here we show that DNA polymerase θ, despite its minor role in chromosomal DNA repair, substantially contributes to random integration, and that cells lacking both DNA polymerase θ and DNA ligase IV, which is essential for non-homologous end joining (NHEJ), exhibit 100% efficiency of spontaneous gene targeting by virtue of undetectable levels of random integration. Thus, DNA polymerase θ-mediated end joining is the sole homology-independent repair route in the absence of NHEJ and, intriguingly, their combined absence reveals rare Alu-Alu recombination events utilizing a stretch of homology. Our findings provide new insights into the mechanics of foreign DNA integration and the role of DNA polymerase θ in human genome maintenance.


Assuntos
Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Linhagem Celular , Humanos , DNA Polimerase teta
6.
FEBS J ; 284(17): 2748-2763, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28622436

RESUMO

Gene targeting by homologous recombination provides the definitive tool for analyzing gene function. Promoterless vectors, which do not possess a promoter to drive marker gene expression, confer higher targeting efficiencies than conventional vectors due to the reduced number of drug-resistant clones. We here show that gene-targeting efficiency is typically ≥ 25% with the use of exon-trapping-type promoterless vectors in a human diploid cell line, Nalm-6. The efficiency of exon-trapping gene targeting was correlated with the level of target gene expression when a 2A peptide sequence was linked to the marker gene. Intriguingly, total arm length was not necessarily a determinant of targeting efficiency, as longer arms tend to enhance both homologous (targeted) and nonhomologous (nontargeted) integration of the vector; rather, the presence of an exon in the 5' arm led to a decreased targeting efficiency. Strikingly, loss of Rad54 did not severely affect the targeting efficiency of exon-trap vectors. Moreover, additional deletion of the Rad54 paralog Rad54B had limited impact on the high-efficiency gene targeting. These results indicate that targeted integration occurs in human cells even when both Rad54 and Rad54B are missing. These studies provide additional important insight into the contribution of various DNA repair factors on the targeting mechanics.


Assuntos
DNA Helicases/genética , Técnicas de Inativação de Genes , Proteínas Nucleares/genética , Linhagem Celular , Proteínas de Ligação a DNA , Éxons , Loci Gênicos , Vetores Genéticos , Humanos , Regiões Promotoras Genéticas
7.
J Biol Chem ; 292(8): 3201-3212, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28082682

RESUMO

Stress-induced activation of p53 is an essential cellular response to prevent aberrant cell proliferation and cancer development. The ubiquitin ligase MDM2 promotes p53 degradation and limits the duration of p53 activation. It remains unclear, however, how p53 persistently escapes MDM2-mediated negative control for making appropriate cell fate decisions. Here we report that TBP-like protein (TLP), a member of the TBP family, is a new regulatory factor for the p53-MDM2 interplay and thus for p53 activation. We found that TLP acts to stabilize p53 protein to ensure long-lasting p53 activation, leading to potentiation of p53-induced apoptosis and senescence after genotoxic stress. Mechanistically, TLP interferes with MDM2 binding and ubiquitination of p53. Moreover, single cell imaging analysis shows that TLP depletion accelerates MDM2-mediated nuclear export of p53. We further show that a cervical cancer-derived TLP mutant has less p53 binding ability and lacks a proliferation-repressive function. Our findings uncover a role of TLP as a competitive MDM2 blocker, proposing a novel mechanism by which p53 escapes the p53-MDM2 negative feedback loop to modulate cell fate decisions.


Assuntos
Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Semelhantes à Proteína de Ligação a TATA-Box/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Células HCT116 , Células HeLa , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas c-mdm2/análise , Proteínas Semelhantes à Proteína de Ligação a TATA-Box/análise , Proteína Supressora de Tumor p53/análise , Ubiquitinação
8.
J Hum Genet ; 61(8): 679-85, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27169690

RESUMO

In successive reports from 2014 to 2015, X-ray repair cross-complementing protein 4 (XRCC4) has been identified as a novel causative gene of primordial dwarfism. XRCC4 is indispensable for non-homologous end joining (NHEJ), the major pathway for repairing DNA double-strand breaks. As NHEJ is essential for V(D)J recombination during lymphocyte development, it is generally believed that abnormalities in XRCC4 cause severe combined immunodeficiency. Contrary to expectations, however, no overt immunodeficiency has been observed in patients with primordial dwarfism harboring XRCC4 mutations. Here, we describe the various XRCC4 mutations that lead to disease and discuss their impact on NHEJ and V(D)J recombination.


Assuntos
Proteínas de Ligação a DNA/genética , Nanismo Hipofisário/genética , Nanismo Hipofisário/imunologia , Estudos de Associação Genética , Mutação , Animais , Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP/metabolismo , Proteínas de Ligação a DNA/metabolismo , Suscetibilidade a Doenças , Nanismo Hipofisário/diagnóstico , Estabilidade Enzimática , Humanos , Imunidade/genética , Síndromes de Imunodeficiência , Fenótipo , Ligação Proteica
9.
Biol Pharm Bull ; 39(1): 25-32, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26725425

RESUMO

Gene targeting via homologous recombination, albeit highly inefficient in human cells, is considered a powerful tool for analyzing gene functions. Despite recent progress in the application of artificial nucleases for genome editing, safety issues remain a concern, particularly when genetic modification is used for therapeutic purposes. Therefore, the development of gene-targeting vectors is necessary for safe and sophisticated genetic modification. In this paper, we describe the effect of vector structure on random integration, which is a major obstacle in efficient gene targeting. In addition, we focus on the features of exon-trapping-type gene-targeting vectors, and discuss a novel strategy for negative selection to enhance gene targeting in human cells.


Assuntos
Regulação da Expressão Gênica/fisiologia , Marcação de Genes/métodos , Engenharia Genética/métodos , Vetores Genéticos , Humanos
10.
BMC Res Notes ; 8: 278, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26123730

RESUMO

BACKGROUND: Targeted gene modification by homologous recombination provides a powerful tool for studying gene function in cells and animals. In higher eukaryotes, non-homologous integration of targeting vectors occurs several orders of magnitude more frequently than does targeted integration, making the gene-targeting technology highly inefficient. For this reason, negative-selection strategies have been employed to reduce the number of drug-resistant clones associated with non-homologous vector integration, particularly when artificial nucleases to introduce a DNA break at the target site are unavailable or undesirable. As such, an exon-trap strategy using a promoterless drug-resistance marker gene provides an effective way to counterselect non-homologous integrants. However, constructing exon-trapping targeting vectors has been a time-consuming and complicated process. RESULTS: By virtue of highly efficient att-mediated recombination, we successfully developed a simple and rapid method to construct plasmid-based vectors that allow for exon-trapping gene targeting. These exon-trap vectors were useful in obtaining correctly targeted clones in mouse embryonic stem cells and human HT1080 cells. Most importantly, with the use of a conditionally cytotoxic gene, we further developed a novel strategy for negative selection, thereby enhancing the efficiency of counterselection for non-homologous integration of exon-trap vectors. CONCLUSIONS: Our methods will greatly facilitate exon-trapping gene-targeting technologies in mammalian cells, particularly when combined with the novel negative selection strategy.


Assuntos
Éxons/genética , Marcação de Genes/métodos , Vetores Genéticos/genética , Animais , Linhagem Celular , Humanos , Camundongos , Células-Tronco
11.
PLoS One ; 9(9): e108236, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25250686

RESUMO

Random integration of targeting vectors into the genome is the primary obstacle in human somatic cell gene targeting. Non-homologous end-joining (NHEJ), a major pathway for repairing DNA double-strand breaks, is thought to be responsible for most random integration events; however, absence of DNA ligase IV (LIG4), the critical NHEJ ligase, does not significantly reduce random integration frequency of targeting vector in human cells, indicating robust integration events occurring via a LIG4-independent mechanism. To gain insights into the mechanism and robustness of LIG4-independent random integration, we employed various types of targeting vectors to examine their integration frequencies in LIG4-proficient and deficient human cell lines. We find that the integration frequency of targeting vector correlates well with the length of homology arms and with the amount of repetitive DNA sequences, especially SINEs, present in the arms. This correlation was prominent in LIG4-deficient cells, but was also seen in LIG4-proficient cells, thus providing evidence that LIG4-independent random integration occurs frequently even when NHEJ is functionally normal. Our results collectively suggest that random integration frequency of conventional targeting vectors is substantially influenced by homology arms, which typically harbor repetitive DNA sequences that serve to facilitate LIG4-independent random integration in human cells, regardless of the presence or absence of functional NHEJ.


Assuntos
DNA Ligases/metabolismo , Marcação de Genes , Linhagem Celular , Reparo do DNA por Junção de Extremidades , DNA Ligase Dependente de ATP , DNA Ligases/genética , Deleção de Genes , Marcação de Genes/métodos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos
12.
PLoS One ; 8(8): e72253, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967291

RESUMO

Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are two major pathways for repairing DNA double-strand breaks (DSBs); however, their respective roles in human somatic cells remain to be elucidated. Here we show using a series of human gene-knockout cell lines that NHEJ repairs nearly all of the topoisomerase II- and low-dose radiation-induced DNA damage, while it negatively affects survival of cells harbouring replication-associated DSBs. Intriguingly, we find that loss of DNA ligase IV, a critical NHEJ ligase, and Artemis, an NHEJ factor with endonuclease activity, independently contribute to increased resistance to replication-associated DSBs. We also show that loss of Artemis alleviates hypersensitivity of DNA ligase IV-null cells to low-dose radiation- and topoisomerase II-induced DSBs. Finally, we demonstrate that Artemis-null human cells display increased gene-targeting efficiencies, particularly in the absence of DNA ligase IV. Collectively, these data suggest that DNA ligase IV and Artemis act cooperatively to promote NHEJ, thereby suppressing HR. Our results point to the possibility that HR can only operate on accidental DSBs when NHEJ is missing or abortive, and Artemis may be involved in pathway switching from incomplete NHEJ to HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , DNA Ligases/metabolismo , Recombinação Homóloga , Proteínas Nucleares/metabolismo , Linhagem Celular , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos da radiação , DNA Ligase Dependente de ATP , DNA Ligases/deficiência , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA , Endonucleases , Técnicas de Inativação de Genes , Marcação de Genes , Recombinação Homóloga/efeitos dos fármacos , Recombinação Homóloga/efeitos da radiação , Humanos , Mutação , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Inibidores da Topoisomerase II/farmacologia
13.
Gene ; 492(1): 305-8, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22119888

RESUMO

Electroporation is a powerful and convenient means for transfection of nonviral vectors into mammalian cells, providing an essential tool for numerous applications including gene targeting via homologous recombination. Recent evidence clearly suggests that high-efficiency gene transfer can be achieved in most cell lines by nucleofection, an electroporation-based transfection method that allows transfected vectors to directly enter the nucleus. In this paper, we analyze the effectiveness of nucleofection for gene targeting using human pre-B cells. For this, we tested 93 different transfection conditions, and found several conditions that gave high (~80%) transfection efficiency with low cytotoxicity (~70% survival rate). Remarkably, under the optimal nucleofection conditions, the gene-targeting efficiency was ~2-5-fold higher than that achieved with conventional electroporation methods. We also found that nucleofection conditions with high transfection efficiency and low cytotoxicity tend to provide high gene-targeting efficiency. Our results provide significant implications for gene targeting, and suggest that nucleofection-based nonviral gene transfer is useful for systematic generation of human gene-knockout cell lines.


Assuntos
Eletroporação/métodos , Marcação de Genes/métodos , Células Precursoras de Linfócitos B , Transfecção , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...