Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(5): 328, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-37198153

RESUMO

It has been well-established that mutations in BRCA1 and BRCA2, compromising functions in DNA double-strand break repair (DSBR), confer hereditary breast and ovarian cancer risk. Importantly, mutations in these genes explain only a minor fraction of the hereditary risk and of the subset of DSBR deficient tumors. Our screening efforts identified two truncating germline mutations in the gene encoding the BRCA1 complex partner ABRAXAS1 in German early-onset breast cancer patients. To unravel the molecular mechanisms triggering carcinogenesis in these carriers of heterozygous mutations, we examined DSBR functions in patient-derived lymphoblastoid cells (LCLs) and in genetically manipulated mammary epithelial cells. By use of these strategies we were able to demonstrate that these truncating ABRAXAS1 mutations exerted dominant effects on BRCA1 functions. Interestingly, we did not observe haploinsufficiency regarding homologous recombination (HR) proficiency (reporter assay, RAD51-foci, PARP-inhibitor sensitivity) in mutation carriers. However, the balance was shifted to use of mutagenic DSBR-pathways. The dominant effect of truncated ABRAXAS1 devoid of the C-terminal BRCA1 binding site can be explained by retention of the N-terminal interaction sites for other BRCA1-A complex partners like RAP80. In this case BRCA1 was channeled from the BRCA1-A to the BRCA1-C complex, which induced single-strand annealing (SSA). Further truncation, additionally deleting the coiled-coil region of ABRAXAS1, unleashed excessive DNA damage responses (DDRs) de-repressing multiple DSBR-pathways including SSA and non-homologous end-joining (NHEJ). Our data reveal de-repression of low-fidelity repair activities as a common feature of cells from patients with heterozygous mutations in genes encoding BRCA1 and its complex partners.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias da Mama/patologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Mutagênese , Mutação
2.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36362151

RESUMO

BRCA1 is a well-known breast cancer risk gene, involved in DNA damage repair via homologous recombination (HR) and replication fork protection. Therapy resistance was linked to loss and amplification of the BRCA1 gene causing inferior survival of breast cancer patients. Most studies have focused on the analysis of complete loss or mutations in functional domains of BRCA1. How mutations in non-functional domains contribute to resistance mechanisms remains elusive and was the focus of this study. Therefore, clones of the breast cancer cell line MCF7 with indels in BRCA1 exon 9 and 14 were generated using CRISPR/Cas9. Clones with successful introduced BRCA1 mutations were evaluated regarding their capacity to perform HR, how they handle DNA replication stress (RS), and the consequences on the sensitivity to MMC, PARP1 inhibition, and ionizing radiation. Unexpectedly, BRCA1 mutations resulted in both increased sensitivity and resistance to exogenous DNA damage, despite a reduction of HR capacity in all clones. Resistance was associated with improved DNA double-strand break repair and reduction in replication stress (RS). Lower RS was accompanied by increased activation and interaction of proteins essential for the S phase-specific DNA damage response consisting of HR proteins, FANCD2, and CHK1.


Assuntos
Neoplasias da Mama , Genes BRCA1 , Humanos , Feminino , Linhagem Celular Tumoral , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Recombinação Homóloga , Reparo do DNA/genética , Replicação do DNA , Dano ao DNA , Neoplasias da Mama/genética , Neoplasias da Mama/tratamento farmacológico
3.
Nucleic Acids Res ; 48(21): 12188-12203, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33166398

RESUMO

We have previously reported that p53 decelerates nascent DNA elongation in complex with the translesion synthesis (TLS) polymerase ι (POLι) which triggers a homology-directed DNA damage tolerance (DDT) pathway to bypass obstacles during DNA replication. Here, we demonstrate that this DDT pathway relies on multiple p53 activities, which can be disrupted by TP53 mutations including those frequently found in cancer tissues. We show that the p53-mediated DDT pathway depends on its oligomerization domain (OD), while its regulatory C-terminus is not involved. Mutation of residues S315 and D48/D49, which abrogate p53 interactions with the DNA repair and replication proteins topoisomerase I and RPA, respectively, and residues L22/W23, which disrupt formation of p53-POLι complexes, all prevent this DDT pathway. Our results demonstrate that the p53-mediated DDT requires the formation of a DNA binding-proficient p53 tetramer, recruitment of such tetramer to RPA-coated forks and p53 complex formation with POLι. Importantly, our mutational analysis demonstrates that transcriptional transactivation is dispensable for the POLι-mediated DDT pathway, which we show protects against DNA replication damage from endogenous and exogenous sources.


Assuntos
Reparo do DNA , DNA Topoisomerases Tipo I/genética , DNA Polimerase Dirigida por DNA/genética , DNA/genética , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , DNA/metabolismo , Dano ao DNA , Replicação do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Células K562 , Mutação , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Oxidantes/farmacologia , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , DNA Polimerase iota
4.
Hum Mol Genet ; 28(24): 4148-4160, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31630195

RESUMO

Whilst heterozygous germline mutations in the ABRAXAS1 gene have been associated with a hereditary predisposition to breast cancer, their effect on promoting tumourigenesis at the cellular level has not been explored. Here, we demonstrate in patient-derived cells that the Finnish ABRAXAS1 founder mutation (c.1082G > A, Arg361Gln), even in the heterozygous state leads to decreased BRCA1 protein levels as well as reduced nuclear localization and foci formation of BRCA1 and CtIP. This causes disturbances in basal BRCA1-A complex localization, which is reflected by a restraint in error-prone DNA double-strand break repair pathway usage, attenuated DNA damage response and deregulated G2-M checkpoint control. The current study clearly demonstrates how the Finnish ABRAXAS1 founder mutation acts in a dominant-negative manner on BRCA1 to promote genome destabilization in heterozygous carrier cells.


Assuntos
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias da Mama/genética , Proteínas de Transporte/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Mutação em Linhagem Germinativa , Adulto , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Feminino , Genes BRCA1 , Predisposição Genética para Doença , Heterozigoto , Humanos , Proteínas Supressoras de Tumor/genética
5.
J Cell Sci ; 132(19)2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31492757

RESUMO

Nucleoli have attracted interest for their role as cellular stress sensors and as potential targets for cancer treatment. The effect of DNA double-strand breaks (DSBs) in nucleoli on rRNA transcription and nucleolar organisation appears to depend on the agent used to introduce DSBs, DSB frequency and the presence (or not) of DSBs outside the nucleoli. To address the controversy, we targeted nucleoli with carbon ions at the ion microbeam SNAKE. Localized ion irradiation with 1-100 carbon ions per point (about 0.3-30 Gy per nucleus) did not lead to overall reduced ribonucleotide incorporation in the targeted nucleolus or other nucleoli of the same cell. However, both 5-ethynyluridine incorporation and Parp1 protein levels were locally decreased at the damaged nucleolar chromatin regions marked by γH2AX, suggesting localized inhibition of rRNA transcription. This locally restricted transcriptional inhibition was not accompanied by nucleolar segregation, a structural reorganisation observed after inhibition of rRNA transcription by treatment with actinomycin D or UV irradiation. The presented data indicate that even multiple complex DSBs do not lead to a pan-nucleolar response if they affect only a subnucleolar region.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/genética , RNA Ribossômico/genética , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , DNA Ribossômico/genética , Humanos , Região Organizadora do Nucléolo/genética , Região Organizadora do Nucléolo/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Transcrição Gênica/genética
6.
Int J Mol Sci ; 19(8)2018 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-30104465

RESUMO

The mammalian DNA replication program is controlled at two phases, the licensing of potential origins of DNA replication in early gap 1 (G1), and the selective firing of a subset of licenced origins in the synthesis (S) phase. Upon entry into the S phase, serine/threonine-protein kinase ATR (ATR) is required for successful completion of the DNA replication program by limiting unnecessary dormant origin activation. Equally important is its activator, DNA topoisomerase 2-binding protein 1 (TopBP1), which is also required for the initiation of DNA replication after a rise in S-phase kinase levels. However, it is unknown how the ATR activation domain of TopBP1 affects DNA replication dynamics. Using human cells conditionally expressing a TopBP1 mutant deficient for ATR activation, we show that functional TopBP1 is required in suppressing local dormant origin activation. Our results demonstrate a regulatory role for TopBP1 in the local balancing of replication fork firing within the S phase.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular Tumoral , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Doxiciclina/farmacologia , Humanos , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Proteínas Nucleares/química , Proteínas Nucleares/genética , Domínios Proteicos/genética , Fase S , Transcrição Gênica/efeitos dos fármacos
7.
Nucleic Acids Res ; 45(6): 3217-3230, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28100698

RESUMO

Cell division cycle protein 45 (Cdc45) is an essential component of the eukaryotic replicative DNA helicase. We found that human Cdc45 forms a complex with the single-stranded DNA (ssDNA) binding protein RPA. Moreover, it actively loads RPA onto nascent ssDNA. Pull-down assays and surface plasmon resonance studies revealed that Cdc45-bound RPA complexed with ssDNA in the 8-10 nucleotide binding mode, but dissociated when RPA covered a 30-mer. Real-time analysis of RPA-ssDNA binding demonstrated that Cdc45 catalytically loaded RPA onto ssDNA. This placement reaction required physical contacts of Cdc45 with the RPA70A subdomain. Our results imply that Cdc45 controlled stabilization of the 8-nt RPA binding mode, the subsequent RPA transition into 30-mer mode and facilitated an ordered binding to ssDNA. We propose that a Cdc45-mediated loading guarantees a seamless deposition of RPA on newly emerging ssDNA at the nascent replication fork.


Assuntos
Proteínas de Ciclo Celular/metabolismo , DNA de Cadeia Simples/metabolismo , Proteína de Replicação A/metabolismo , Sítios de Ligação , Proteínas de Ciclo Celular/química , Humanos , Modelos Moleculares , Ligação Proteica , Proteína de Replicação A/química
8.
FEBS Lett ; 590(23): 4233-4241, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27805738

RESUMO

High fidelity of genome duplication is ensured by cooperation of polymerase proofreading and mismatch repair (MMR) activities. Here, we show that human mismatch recognizing proteins MutS homolog 2 (MSH2) and MSH6 copurify and interact with replicative Pol α. This enzyme also is the replicative primase and replicates DNA with poor fidelity. We show that MSH2 associates with known human replication origins with different dynamics than DNA polymerase (Pol α). Furthermore, we explored the potential functional role of Pol α in the mismatch repair reaction using an in vitro mismatch repair assay and observed that Pol α promotes mismatch repair. Taken together, we show that human Pol α interacts with MSH2-MSH6 complex and propose that this interaction occurs during the mismatch repair reaction.


Assuntos
Reparo de Erro de Pareamento de DNA , DNA Polimerase I/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Replicação do DNA , Células HeLa , Humanos , Ligação Proteica , Especificidade por Substrato
9.
Oncotarget ; 7(51): 85365-85380, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27863419

RESUMO

Tuberous Sclerosis (TSC) is characterized by exorbitant mTORC1 signalling and manifests as non-malignant, apoptosis-prone neoplasia. Previous reports have shown that TSC-/- cells are highly susceptible to mild, innocuous doses of genotoxic stress, which drive TSC-/- cells into apoptotic death. It has been argued that this hypersensitivity to stress derives from a metabolic/energetic shortfall in TSC-/- cells, but how metabolic dysregulation affects the DNA damage response and cell cycle alterations in TSC-/- cells exposed to genotoxic stress is not understood. We report here the occurrence of futile checkpoint responses and an unusual type of replicative stress (RS) in TSC1-/- fibroblasts exposed to low-dose genotoxins. This RS is characterized by elevated nucleotide incorporation rates despite only modest origin over-firing. Strikingly, an increased propensity for asymmetric fork progression and profuse chromosomal aberrations upon mild DNA damage confirmed that TSC loss indeed proved detrimental to stress adaptation. We conclude that low stress tolerance of TSC-/- cells manifests at the level of DNA replication control, imposing strong negative selection on genomic instability that could in turn detain TSC-mutant tumours benign.


Assuntos
Dano ao DNA , Replicação do DNA , Doxorrubicina/toxicidade , Fibroblastos/efeitos dos fármacos , Hidroxiureia/toxicidade , Sirolimo/toxicidade , Proteínas Supressoras de Tumor/deficiência , Animais , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Aberrações Cromossômicas/induzido quimicamente , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Fibroblastos/patologia , Instabilidade Genômica/efeitos dos fármacos , Camundongos Knockout , Interferência de RNA , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transfecção , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
10.
Cell Cycle ; 15(20): 2766-79, 2016 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-27590262

RESUMO

The repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) is an essential process in maintenance of chromosomal stability. A key player of HR is the strand exchange factor RAD51 whose assembly at sites of DNA damage is tightly regulated. We detected an endogenous complex of RAD51 with the calcium-binding protein S100A11, which is localized at sites of DNA repair in HaCaT cells as well as in normal human epidermal keratinocytes (NHEK) synchronized in S phase. In biochemical assays, we revealed that S100A11 enhanced the RAD51 strand exchange activity. When cells expressing a S100A11 mutant lacking the ability to bind Ca(2+), a prolonged persistence of RAD51 in repair sites and nuclear γH2AX foci was observed suggesting an incomplete DNA repair. The same phenotype became apparent when S100A11 was depleted by RNA interference. Furthermore, down-regulation of S100A11 resulted in both reduced sister chromatid exchange confirming the restriction of the recombination capacity of the cells, and in an increase of chromosomal aberrations reflecting the functional requirement of S100A11 for the maintenance of genomic stability. Our data indicate that S100A11 is involved in homologous recombination by regulating the appearance of RAD51 in DSB repair sites. This function requires the calcium-binding activity of S100A11.


Assuntos
Reparo do DNA/genética , Genoma Humano , Recombinação Homóloga/genética , Rad51 Recombinase/metabolismo , Proteínas S100/metabolismo , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular , Aberrações Cromossômicas , Dano ao DNA/genética , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Proteínas Mutantes/metabolismo , Ligação Proteica/genética
11.
Proc Natl Acad Sci U S A ; 113(30): E4311-9, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27407148

RESUMO

DNA damage tolerance facilitates the progression of replication forks that have encountered obstacles on the template strands. It involves either translesion DNA synthesis initiated by proliferating cell nuclear antigen monoubiquitination or less well-characterized fork reversal and template switch mechanisms. Herein, we characterize a novel tolerance pathway requiring the tumor suppressor p53, the translesion polymerase ι (POLι), the ubiquitin ligase Rad5-related helicase-like transcription factor (HLTF), and the SWI/SNF catalytic subunit (SNF2) translocase zinc finger ran-binding domain containing 3 (ZRANB3). This novel p53 activity is lost in the exonuclease-deficient but transcriptionally active p53(H115N) mutant. Wild-type p53, but not p53(H115N), associates with POLι in vivo. Strikingly, the concerted action of p53 and POLι decelerates nascent DNA elongation and promotes HLTF/ZRANB3-dependent recombination during unperturbed DNA replication. Particularly after cross-linker-induced replication stress, p53 and POLι also act together to promote meiotic recombination enzyme 11 (MRE11)-dependent accumulation of (phospho-)replication protein A (RPA)-coated ssDNA. These results implicate a direct role of p53 in the processing of replication forks encountering obstacles on the template strand. Our findings define an unprecedented function of p53 and POLι in the DNA damage response to endogenous or exogenous replication stress.


Assuntos
Dano ao DNA , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , DNA/química , DNA/genética , DNA/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/genética , Recombinação Homóloga , Humanos , Células K562 , Conformação de Ácido Nucleico , Interferência de RNA , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética , DNA Polimerase iota
12.
Cell Cycle ; 15(7): 974-85, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26919204

RESUMO

Cdc45 is an essential protein that together with Mcm2-7 and GINS forms the eukaryotic replicative helicase CMG. Cdc45 seems to be rate limiting for the initial unwinding or firing of replication origins. In line with this view, Cdc45-overexpressing cells fired at least twice as many origins as control cells. However, these cells displayed an about 2-fold diminished fork elongation rate, a pronounced asymmetry of replication fork extension, and an early S phase arrest. This was accompanied by H2AX-phosphorylation and subsequent apoptosis. Unexpectedly, we did not observe increased ATR/Chk1 signaling but rather a mild ATM/Chk2 response. In addition, we detected accumulation of long stretches of single-stranded DNA, a hallmark of replication catastrophe. We conclude that increased origin firing by upregulated Cdc45 caused exhaustion of the single-strand binding protein RPA, which in consequence diminished the ATR/Chk1 response; the subsequently occurring fork breaks led to an ATM/Chk2 mediated phosphorylation of H2AX and eventually to apoptosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Apoptose , DNA de Cadeia Simples/análise , Células HeLa , Histonas/metabolismo , Humanos , Origem de Replicação , Pontos de Checagem da Fase S do Ciclo Celular , Transdução de Sinais
13.
Cell Cycle ; 14(19): 3190-202, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317153

RESUMO

In response to replication stress ATR signaling through CHK1 controls the intra-S checkpoint and is required for the maintenance of genomic integrity. Homologous recombination (HR) comprises a series of interrelated pathways that function in the repair of DNA double strand breaks and interstrand crosslinks. In addition, HR, with its key player RAD51, provides critical support for the recovery of stalled forks during replication. High levels of RAD51 are regularly found in various cancers, yet little is known about the effect of the increased RAD51 expression on intra-S checkpoint signaling. Here, we describe a role for RAD51 in driving genomic instability caused by impaired replication and intra-S mediated CHK1 signaling by studying an inducible RAD51 overexpression model as well as 10 breast cancer cell lines. We demonstrate that an excess of RAD51 decreases I-Sce-I mediated HR despite formation of more RAD51 foci. Cells with high RAD51 levels display reduced elongation rates and excessive dormant origin firing during undisturbed growth and after damage, likely caused by impaired CHK1 activation. In consequence, the inability of cells with a surplus of RAD51 to properly repair complex DNA damage and to resolve replication stress leads to higher genomic instability and thus drives tumorigenesis.


Assuntos
Proteínas Quinases/metabolismo , Rad51 Recombinase/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quebras de DNA de Cadeia Dupla , Dano ao DNA/genética , Dano ao DNA/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , Replicação do DNA/genética , Replicação do DNA/fisiologia , Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Recombinação Homóloga/genética , Recombinação Homóloga/fisiologia , Humanos , Proteínas Quinases/genética , Rad51 Recombinase/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
14.
Nucleic Acids Res ; 43(10): 4975-89, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25916852

RESUMO

Nucleoli are not only organelles that produce ribosomal subunits. They are also overarching sensors of different stress conditions and they control specific nucleolar stress pathways leading to stabilization of p53. During DNA replication, ATR and its activator TopBP1 initiate DNA damage response upon DNA damage and replication stress. We found that a basal level of TopBP1 protein associates with ribosomal DNA repeat. When upregulated, TopBP1 concentrates at the ribosomal chromatin and initiates segregation of nucleolar components--the hallmark of nucleolar stress response. TopBP1-induced nucleolar segregation is coupled to shut-down of ribosomal RNA transcription in an ATR-dependent manner. Nucleolar segregation induced by TopBP1 leads to a moderate elevation of p53 protein levels and to localization of activated p53 to nucleolar caps containing TopBP1, UBF and RNA polymerase I. Our findings demonstrate that TopBP1 and ATR are able to inhibit the synthesis of rRNA and to activate nucleolar stress pathway; yet the p53-mediated cell cycle arrest is thwarted in cells expressing high levels of TopBP1. We suggest that inhibition of rRNA transcription by different stress regulators is a general mechanism for cells to initiate nucleolar stress pathway.


Assuntos
Proteínas de Transporte/metabolismo , Nucléolo Celular/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , RNA Ribossômico/biossíntese , Transcrição Gênica , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Transporte/química , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Nucléolo Celular/enzimologia , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , DNA Ribossômico/química , Proteínas de Ligação a DNA/química , Humanos , Proteínas Nucleares/química , Estrutura Terciária de Proteína , RNA Ribossômico/genética , Sequências Repetitivas de Ácido Nucleico
15.
Nucleic Acids Res ; 42(20): 12614-27, 2014 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-25336622

RESUMO

Human RecQL4 belongs to the ubiquitous RecQ helicase family. Its N-terminal region represents the only homologue of the essential DNA replication initiation factor Sld2 of Saccharomyces cerevisiae, and also participates in the vertebrate initiation of DNA replication. Here, we utilized a random screen to identify N-terminal fragments of human RecQL4 that could be stably expressed in and purified from Escherichia coli. Biophysical characterization of these fragments revealed that the Sld2 homologous RecQL4 N-terminal domain carries large intrinsically disordered regions. The N-terminal fragments were sufficient for the strong annealing activity of RecQL4. Moreover, this activity appeared to be the basis for an ATP-independent strand exchange activity. Both activities relied on multiple DNA-binding sites with affinities to single-stranded, double-stranded and Y-structured DNA. Finally, we found a remarkable affinity of the N-terminus for guanine quadruplex (G4) DNA, exceeding the affinities for other DNA structures by at least 60-fold. Together, these findings suggest that the DNA interactions mediated by the N-terminal region of human RecQL4 represent a central function at the replication fork. The presented data may also provide a mechanistic explanation for the role of elements with a G4-forming propensity identified in the vicinity of vertebrate origins of DNA replication.


Assuntos
DNA/metabolismo , RecQ Helicases/química , RecQ Helicases/metabolismo , Sítios de Ligação , DNA/química , Proteínas de Ligação a DNA/química , Quadruplex G , Humanos , Proteínas Intrinsicamente Desordenadas/química , Ligação Proteica , Estrutura Terciária de Proteína
16.
PLoS One ; 9(4): e93908, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24710081

RESUMO

DNA polymerase ε (pol ε) is believed to be the leading strand replicase in eukaryotes whereas pols λ and ß are thought to be mainly involved in re-synthesis steps of DNA repair. DNA elongation by the human pol ε is halted by an abasic site (apurinic/apyrimidinic (AP) site). We have previously reported that human pols λ, ß and η can perform translesion synthesis (TLS) of an AP site in the presence of pol ε. In the case of pol λ and ß, this TLS requires the presence of a gap downstream from the product synthetized by the ε replicase. However, since these studies were conducted exclusively with a linear DNA template, we decided to test whether the structure of the template could influence the capacity of the pols ε, λ, ß and η to perform TLS of an AP site. Therefore, we have investigated the replication of damaged "minicircle" DNA templates. In addition, replication of circular DNA requires, beyond DNA pols, the processivity clamp PCNA, the clamp loader replication factor C (RFC), and the accessory proteins replication protein A (RPA). Finally we have compared the capacity of unmodified versus monoubiquitinated PCNA in sustaining TLS by pols λ and η on a circular template. Our results indicate that in vitro gap-directed TLS synthesis by pols λ and ß in the presence of pol ε, RPA and PCNA is unaffected by the structure of the DNA template. Moreover, monoubiquitination of PCNA does not affect TLS by pol λ while it appears to slightly stimulate TLS by pol η.


Assuntos
DNA Polimerase II/metabolismo , Replicação do DNA , DNA Circular , DNA Polimerase II/genética , DNA Polimerase beta/genética , DNA Polimerase beta/metabolismo , Reparo do DNA , Humanos
17.
Nucleic Acids Res ; 42(4): 2308-19, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24293646

RESUMO

The cell division cycle protein 45 (Cdc45) represents an essential replication factor that, together with the Mcm2-7 complex and the four subunits of GINS, forms the replicative DNA helicase in eukaryotes. Recombinant human Cdc45 (hCdc45) was structurally characterized and its DNA-binding properties were determined. Synchrotron radiation circular dichroism spectroscopy, dynamic light scattering, small-angle X-ray scattering and atomic force microscopy revealed that hCdc45 exists as an alpha-helical monomer and possesses a structure similar to its bacterial homolog RecJ. hCdc45 bound long (113-mer or 80-mer) single-stranded DNA fragments with a higher affinity than shorter ones (34-mer). hCdc45 displayed a preference for 3' protruding strands and bound tightly to single-strand/double-strand DNA junctions, such as those presented by Y-shaped DNA, bubbles and displacement loops, all of which appear transiently during the initiation of DNA replication. Collectively, our findings suggest that hCdc45 not only binds to but also slides on DNA with a 3'-5' polarity and, thereby acts as a molecular 'wedge' to initiate DNA strand displacement.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ligação a DNA/química , DNA/metabolismo , Proteínas de Ciclo Celular/metabolismo , DNA/química , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Conformação de Ácido Nucleico , Ligação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína
18.
Nat Commun ; 4: 2578, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24153426

RESUMO

Besides mutations in BRCA1/BRCA2, heterozygous defects in PALB2 are important in breast cancer predisposition. PALB2 heterozygosity increases the risk of malignancy about sixfold. PALB2 interacts with BRCA1 and BRCA2 to regulate homologous recombination and mediate DNA damage response. Here we show, by analysing lymphoblastoid cell lines from heterozygous female PALB2 mutation carriers, that PALB2 haploinsufficiency causes aberrant DNA replication/damage response. Mutation carrier cells show increased origin firing and shorter distance between consecutive replication forks. Carrier cell lines also show elevated ATR protein, but not phosphorylation levels, and a majority of them display aberrant Chk1-/Chk2-mediated DNA damage response. Elevated chromosome instability is observed in primary blood lymphocytes of PALB2 mutation carriers, indicating that the described mechanisms of genome destabilization operate also at the organism level. These findings provide a new mechanism for early stages of breast cancer development that may also apply to other heterozygous homologous recombination signalling pathway gene mutations in hereditary cancer predisposition.


Assuntos
Neoplasias da Mama/genética , Replicação do DNA , Regulação Neoplásica da Expressão Gênica , Heterozigoto , Mutação , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Estudos de Casos e Controles , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Instabilidade Cromossômica , Dano ao DNA , Proteína do Grupo de Complementação N da Anemia de Fanconi , Feminino , Predisposição Genética para Doença , Haploinsuficiência , Humanos , Linfócitos/metabolismo , Linfócitos/patologia , Proteínas Nucleares/metabolismo , Cultura Primária de Células , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo
19.
PLoS One ; 7(11): e50045, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209641

RESUMO

Lysyl hydroxylase 3 (LH3) has lysyl hydroxylase, galactosyltransferase, and glucosyltransferase activities, which are sequentially required for the formation of glucosylgalactosyl hydroxylysines in collagens. Here we demonstrate for the first time that LH3 also modifies the lysine residues in the collagenous domain of adiponectin, which has important roles in glucose and lipid metabolism and inflammation. Hydroxylation and, especially, glycosylation of the lysine residues of adiponectin have been shown to be essential for the formation of the more active high molecular weight adiponectin oligomers and thus for its function. In cells that totally lack LH3 enzyme, the galactosylhydroxylysine residues of adiponectin were not glucosylated to glucosylgalactosylhydroxylysine residues and the formation of high and middle molecular weight adiponectin oligomers was impaired. Circulating adiponectin levels in mutant mice lacking the lysyl hydroxylase activity of LH3 were significantly reduced, which indicates that LH3 is required for complete modification of lysine residues in adiponectin and the loss of some of the glycosylated hydroxylysine residues severely affects the secretion of adiponectin. LH mutant mice with reduced adiponectin level showed a high fat diet-induced increase in glucose, triglyceride, and LDL-cholesterol levels, hallmarks of the metabolic syndrome in humans. Our results reveal the first indication that LH3 is an important regulator of adiponectin biosynthesis, secretion and activity and thus might be a potential candidate for therapeutic applications in diseases associated with obesity and insulin resistance.


Assuntos
Adiponectina/química , Adiponectina/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Multimerização Proteica , Sequência de Aminoácidos , Animais , Linhagem Celular , Ativação Enzimática , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Genótipo , Glicosilação , Hidroxilação , Lisina/química , Lisina/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Peso Molecular , Mutação , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
J Biol Chem ; 287(40): 33327-38, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22887995

RESUMO

DNA polymerases (Pol) α, δ, and ε replicate the bulk of chromosomal DNA in eukaryotic cells, Pol ε being the main leading strand and Pol δ the lagging strand DNA polymerase. By applying chromatin immunoprecipitation (ChIP) and quantitative PCR we found that at G(1)/S arrest, all three DNA polymerases were enriched with DNA containing the early firing lamin B2 origin of replication and, 2 h after release from the block, with DNA containing the origin at the upstream promoter region of the MCM4 gene. Pol α, δ, and ε were released from these origins upon firing. All three DNA polymerases, Mcm3 and Cdc45, but not Orc2, still formed complexes in late S phase. Reciprocal ChIP of the three DNA polymerases revealed that at G(1)/S arrest and early in S phase, Pol α, δ, and ε were associated with the same nucleoprotein complexes, whereas in late S phase Pol ε and Pol α/δ were largely associated with distinct complexes. At G(1)/S arrest, the replicative DNA polymerases were associated with lamins, but in late S phase only Pol ε, not Pol α/δ, remained associated with lamins. Consistently, Pol ε, but not Pol δ, was found in nuclear matrix fraction throughout the cell cycle. Therefore, Pol ε and Pol α/δ seem to pursue their functions at least in part independently in late S phase, either by physical uncoupling of lagging strand maturation from the fork progression, or by recruitment of Pol δ, but not Pol ε, to post-replicative processes such as translesion synthesis or post-replicative repair.


Assuntos
DNA Polimerase III/química , DNA Polimerase II/química , DNA Polimerase I/química , Laminas/metabolismo , Catálise , Ciclo Celular , Imunoprecipitação da Cromatina , Replicação do DNA , Regulação da Expressão Gênica , Células HeLa , Humanos , Nucleoproteínas/química , Reação em Cadeia da Polimerase/métodos , Fase S , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...