Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(5)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673424

RESUMO

Telomerase negative cancer cell types use the Alternative Lengthening of Telomeres (ALT) pathway to elongate telomeres ends. Here, we show that silencing human DNA polymerase (Pol λ) in ALT cells represses ALT activity and induces telomeric stress. In addition, replication stress in the absence of Pol λ, strongly affects the survival of ALT cells. In vitro, Pol λ can promote annealing of even a single G-rich telomeric repeat to its complementary strand and use it to prime DNA synthesis. The noncoding telomeric repeat containing RNA TERRA and replication protein A negatively regulate this activity, while the Protection of Telomeres protein 1 (POT1)/TPP1 heterodimer stimulates Pol λ. Pol λ associates with telomeres and colocalizes with TPP1 in cells. In summary, our data suggest a role of Pol λ in the maintenance of telomeres by the ALT mechanism.


Assuntos
Aminopeptidases/metabolismo , DNA Polimerase beta/metabolismo , Quadruplex G , Serina Proteases/metabolismo , Homeostase do Telômero , Proteínas de Ligação a Telômeros/metabolismo , Linhagem Celular Tumoral , Humanos , Complexos Multiproteicos , Proteína de Replicação A/metabolismo , Complexo Shelterina , Telômero/química , Telômero/metabolismo
2.
Nucleic Acids Res ; 47(21): 11268-11283, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31586398

RESUMO

Accurate DNA replication is critical for the maintenance of genome integrity and cellular survival. Cancer-associated alterations often involve key players of DNA replication and of the DNA damage-signalling cascade. Post-translational modifications play a fundamental role in coordinating replication and repair and central among them is ubiquitylation. We show that the E3 ligase UBR5 interacts with components of the replication fork, including the translesion synthesis (TLS) polymerase polη. Depletion of UBR5 leads to replication problems, such as slower S-phase progression, resulting in the accumulation of single stranded DNA. The effect of UBR5 knockdown is related to a mis-regulation in the pathway that controls the ubiquitylation of histone H2A (UbiH2A) and blocking this modification is sufficient to rescue the cells from replication problems. We show that the presence of polη is the main cause of replication defects and cell death when UBR5 is silenced. Finally, we unveil a novel interaction between polη and H2A suggesting that UbiH2A could be involved in polη recruitment to the chromatin and the regulation of TLS.


Assuntos
Dano ao DNA , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Células Cultivadas , Dano ao DNA/genética , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , DNA Polimerase Dirigida por DNA/genética , Histonas/metabolismo , Humanos , Ligação Proteica , Processamento de Proteína Pós-Traducional , Fase S/genética , Ubiquitinação/fisiologia
3.
Nucleic Acids Res ; 45(16): 9441-9454, 2017 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-28934491

RESUMO

DNA translesion synthesis (TLS) is a crucial damage tolerance pathway that oversees the completion of DNA replication in the presence of DNA damage. TLS polymerases are capable of bypassing a distorted template but they are generally considered inaccurate and they need to be tightly regulated. We have previously shown that polη is phosphorylated on Serine 601 after DNA damage and we have demonstrated that this modification is important for efficient damage bypass. Here we report that polη is also phosphorylated by CDK2, in the absence of damage, in a cell cycle-dependent manner and we identify serine 687 as an important residue targeted by the kinase. We discover that phosphorylation on serine 687 regulates the stability of the polymerase during the cell cycle, allowing it to accumulate in late S and G2 when productive TLS is critical for cell survival. Furthermore, we show that alongside the phosphorylation of S601, the phosphorylation of S687 and S510, S512 and/or S514 are important for damage bypass and cell survival after UV irradiation. Taken together our results provide new insights into how cells can, at different times, modulate DNA TLS for improved cell survival.


Assuntos
Ciclo Celular/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Ciclo Celular/efeitos da radiação , Linhagem Celular , Sobrevivência Celular , Quinase 2 Dependente de Ciclina/metabolismo , Dano ao DNA/efeitos da radiação , Reparo do DNA , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , Humanos , Fosforilação , Estabilidade Proteica , Serina/metabolismo , Raios Ultravioleta
4.
Nucleic Acids Res ; 45(5): 2600-2614, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-27994034

RESUMO

Ribonucleotides (rNs) incorporated in the genome by DNA polymerases (Pols) are removed by RNase H2. Cytidine and guanosine preferentially accumulate over the other rNs. Here we show that human Pol η can incorporate cytidine monophosphate (rCMP) opposite guanine, 8-oxo-7,8-dihydroguanine, 8-methyl-2΄-deoxyguanosine and a cisplatin intrastrand guanine crosslink (cis-PtGG), while it cannot bypass a 3-methylcytidine or an abasic site with rNs as substrates. Pol η is also capable of synthesizing polyribonucleotide chains, and its activity is enhanced by its auxiliary factor DNA Pol δ interacting protein 2 (PolDIP2). Human RNase H2 removes cytidine and guanosine less efficiently than the other rNs and incorporation of rCMP opposite DNA lesions further reduces the efficiency of RNase H2. Experiments with XP-V cell extracts indicate Pol η as the major basis of rCMP incorporation opposite cis-PtGG. These results suggest that translesion synthesis by Pol η can contribute to the accumulation of rCMP in the genome, particularly opposite modified guanines.


Assuntos
Dano ao DNA , Reparo do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Ribonuclease H/metabolismo , Ribonucleotídeos/metabolismo , Linhagem Celular , Monofosfato de Citidina/metabolismo , DNA/biossíntese , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , RNA/biossíntese , Xeroderma Pigmentoso/genética
5.
Front Genet ; 7: 105, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27379156

RESUMO

DNA replication is an extremely complex process that needs to be executed in a highly accurate manner in order to propagate the genome. This task requires the coordination of a number of enzymatic activities and it is fragile and prone to arrest after DNA damage. DNA damage tolerance provides a last line of defense that allows completion of DNA replication in the presence of an unrepaired template. One of such mechanisms is called post-replication repair (PRR) and it is used by the cells to bypass highly distorted templates caused by damaged bases. PRR is extremely important for the cellular life and performs the bypass of the damage both in an error-free and in an error-prone manner. In light of these two possible outcomes, PRR needs to be tightly controlled in order to prevent the accumulation of mutations leading ultimately to genome instability. Post-translational modifications of PRR proteins provide the framework for this regulation with ubiquitylation and SUMOylation playing a pivotal role in choosing which pathway to activate, thus controlling the different outcomes of damage bypass. The proliferating cell nuclear antigen (PCNA), the DNA clamp for replicative polymerases, plays a central role in the regulation of damage tolerance and its modification by ubiquitin, and SUMO controls both the error-free and error-prone branches of PRR. Furthermore, a significant number of polymerases are involved in the bypass of DNA damage possess domains that can bind post-translational modifications and they are themselves target for ubiquitylation. In this review, we will focus on how ubiquitin and ubiquitin-like modifications can regulate the DNA damage tolerance systems and how they control the recruitment of different proteins to the replication fork.

6.
Nat Commun ; 7: 10805, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26917111

RESUMO

Oxidative stress is a very frequent source of DNA damage. Many cellular DNA polymerases (Pols) can incorporate ribonucleotides (rNMPs) during DNA synthesis. However, whether oxidative stress-triggered DNA repair synthesis contributes to genomic rNMPs incorporation is so far not fully understood. Human specialized Pols ß and λ are the important enzymes involved in the oxidative stress tolerance, acting both in base excision repair and in translesion synthesis past the very frequent oxidative lesion 7,8-dihydro-8-oxoguanine (8-oxo-G). We found that Pol ß, to a greater extent than Pol λ can incorporate rNMPs opposite normal bases or 8-oxo-G, and with a different fidelity. Further, the incorporation of rNMPs opposite 8-oxo-G delays repair by DNA glycosylases. Studies in Pol ß- and λ-deficient cell extracts suggest that Pol ß levels can greatly affect rNMP incorporation opposite oxidative DNA lesions.


Assuntos
Dano ao DNA , DNA Glicosilases/metabolismo , DNA Polimerase beta/metabolismo , Reparo do DNA , Estresse Oxidativo , Ribonucleotídeos/metabolismo , Animais , Linhagem Celular , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Camundongos
7.
Nucleic Acids Res ; 41(1): 229-41, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23118481

RESUMO

Human DNA polymerase (pol) λ functions in base excision repair and non-homologous end joining. We have previously shown that DNA pol λ is involved in accurate bypass of the two frequent oxidative lesions, 7,8-dihydro-8-oxoguanine and 1,2-dihydro-2-oxoadenine during the S phase. However, nothing is known so far about the relationship of DNA pol λ with the S phase DNA damage response checkpoint. Here, we show that a knockdown of DNA pol λ, but not of its close homologue DNA pol ß, results in replication fork stress and activates the S phase checkpoint, slowing S phase progression in different human cancer cell lines. We furthermore show that DNA pol λ protects cells from oxidative DNA damage and also functions in rescuing stalled replication forks. Its absence becomes lethal for a cell when a functional checkpoint is missing, suggesting a DNA synthesis deficiency. Our results provide the first evidence, to our knowledge, that DNA pol λ is required for cell cycle progression and is functionally connected to the S phase DNA damage response machinery in cancer cells.


Assuntos
DNA Polimerase beta/fisiologia , Replicação do DNA , Pontos de Checagem da Fase S do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Quinase 1 do Ponto de Checagem , Dano ao DNA , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/metabolismo , Células HeLa , Humanos , Hidroxiureia/farmacologia , Estresse Oxidativo , Proteínas Quinases/metabolismo , Interferência de RNA , Reparo de DNA por Recombinação , Estresse Fisiológico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA