Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 643, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36746968

RESUMO

Interstitial lung diseases such as idiopathic pulmonary fibrosis (IPF) are caused by persistent micro-injuries to alveolar epithelial tissues accompanied by aberrant repair processes. IPF is currently treated with pirfenidone and nintedanib, compounds which slow the rate of disease progression but fail to target underlying pathophysiological mechanisms. The DNA repair protein 8-oxoguanine DNA glycosylase-1 (OGG1) has significant roles in the modulation of inflammation and metabolic syndromes. Currently, no pharmaceutical solutions targeting OGG1 have been utilized in the treatment of IPF. In this study we show Ogg1-targeting siRNA mitigates bleomycin-induced pulmonary fibrosis in male mice, highlighting OGG1 as a tractable target in lung fibrosis. The small molecule OGG1 inhibitor, TH5487, decreases myofibroblast transition and associated pro-fibrotic gene expressions in fibroblast cells. In addition, TH5487 decreases levels of pro-inflammatory mediators, inflammatory cell infiltration, and lung remodeling in a murine model of bleomycin-induced pulmonary fibrosis conducted in male C57BL6/J mice. OGG1 and SMAD7 interact to induce fibroblast proliferation and differentiation and display roles in fibrotic murine and IPF patient lung tissue. Taken together, these data suggest that TH5487 is a potentially clinically relevant treatment for IPF but further study in human trials is required.


Assuntos
DNA Glicosilases , Fibrose Pulmonar Idiopática , Pneumonia , Masculino , Camundongos , Humanos , Animais , Pulmão/patologia , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/metabolismo , Fibrose , Pneumonia/metabolismo , Bleomicina/toxicidade , DNA Glicosilases/genética , DNA Glicosilases/metabolismo
7.
Oncogenesis ; 5(12): e280, 2016 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-27991914

RESUMO

Recent whole-exome sequencing of malignancies have detected recurrent somatic mutations in U2 small nuclear ribonucleoprotein complex (snRNP) components of the spliceosome. These factors have also been identified as novel players in the DNA-damage response (DDR) in several genome-wide screens and proteomic analysis. Although accumulating evidence implies that the spliceosome has an important role in genome stability and is an emerging hallmark of cancer, its precise role in DNA repair still remains elusive. Here we identify two distinct mechanisms of how spliceosome U2 snRNP factors contribute to genome stability. We show that the spliceosome maintains protein levels of essential repair factors, thus contributing to homologous recombination repair. In addition, real-time laser microirradiation analysis identified rapid recruitment of the U2 snRNP factor SNRPA1 to DNA-damage sites. Functional analysis of SNRPA1 revealed a more immediate and direct role in preventing R-loop-induced DNA damage. Our present study implies a complex interrelation between transcription, mRNA splicing and the DDR. Cells require rapid spatio-temporal coordination of these chromatin transactions to cope with various forms of genotoxic stress.

8.
Oncogenesis ; 5(12): e275, 2016 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-27918552

RESUMO

hMTH1 is an 8-oxodGTPase that prevents mis-incorporation of free oxidized nucleotides into genomic DNA. Base excision and mismatch repair pathways also restrict the accumulation of oxidized lesions in DNA by removing the mis-inserted 8-oxo-7,8-dihydro-2'-deoxyguanosines (8-oxodGs). In this study, we aimed to investigate the interplay between hMYH DNA glycosylase and hMTH1 for cancer cell survival by using mismatch repair defective T-cell acute lymphoblastic leukemia (T-ALL) cells. To this end, MYH and MTH1 were silenced individually or simultaneously using small hairpin RNAs. Increased sub-G1 population and apoptotic cells were observed upon concurrent depletion of both enzymes. Elevated cell death was consistent with cleaved caspase 3 accumulation in double knockdown cells. Importantly, overexpression of the nuclear isoform of hMYH could remove the G1 arrest and partially rescue the toxicity observed in hMTH1-depleted cells. In addition, expression profiles of human DNA glycosylases were generated using quantitative reverse transcriptase-PCR in MTH1 and/or MYH knockdown cells. NEIL1 DNA glycosylase, involved in repair of oxidized nucleosides, was found to be significantly downregulated as a cellular response to MTH1-MYH co-suppression. Overall, the results suggest that hMYH and hMTH1 functionally cooperate for effective repair and survival in mismatch repair defective T-ALL Jurkat A3 cells.

9.
Ann Oncol ; 27(12): 2275-2283, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27827301

RESUMO

BACKGROUND: Previously, we showed cancer cells rely on the MTH1 protein to prevent incorporation of otherwise deadly oxidised nucleotides into DNA and we developed MTH1 inhibitors which selectively kill cancer cells. Recently, several new and potent inhibitors of MTH1 were demonstrated to be non-toxic to cancer cells, challenging the utility of MTH1 inhibition as a target for cancer treatment. MATERIAL AND METHODS: Human cancer cell lines were exposed in vitro to MTH1 inhibitors or depleted of MTH1 by siRNA or shRNA. 8-oxodG was measured by immunostaining and modified comet assay. Thermal Proteome profiling, proteomics, cellular thermal shift assays, kinase and CEREP panel were used for target engagement, mode of action and selectivity investigations of MTH1 inhibitors. Effect of MTH1 inhibition on tumour growth was explored in BRAF V600E-mutated malignant melanoma patient derived xenograft and human colon cancer SW480 and HCT116 xenograft models. RESULTS: Here, we demonstrate that recently described MTH1 inhibitors, which fail to kill cancer cells, also fail to introduce the toxic oxidized nucleotides into DNA. We also describe a new MTH1 inhibitor TH1579, (Karonudib), an analogue of TH588, which is a potent, selective MTH1 inhibitor with good oral availability and demonstrates excellent pharmacokinetic and anti-cancer properties in vivo. CONCLUSION: We demonstrate that in order to kill cancer cells MTH1 inhibitors must also introduce oxidized nucleotides into DNA. Furthermore, we describe TH1579 as a best-in-class MTH1 inhibitor, which we expect to be useful in order to further validate the MTH1 inhibitor concept.


Assuntos
Enzimas Reparadoras do DNA/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Pirimidinas/administração & dosagem , 8-Hidroxi-2'-Desoxiguanosina , Animais , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/isolamento & purificação , Desoxiguanosina/metabolismo , Humanos , Camundongos , Neoplasias/genética , Neoplasias/patologia , Nucleotídeos/metabolismo , Oxirredução , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , RNA Interferente Pequeno/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cell Death Dis ; 7: e2267, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27310875

RESUMO

Altered expression of the multifunctional protein WRAP53ß (WD40 encoding RNA Antisense to p53), which targets repair factors to DNA double-strand breaks and factors involved in telomere elongation to Cajal bodies, is linked to carcinogenesis. While loss of WRAP53ß function has been shown to disrupt processes regulated by this protein, the consequences of its overexpression remain unclear. Here we demonstrate that overexpression of WRAP53ß disrupts the formation of and impairs the localization of coilin to Cajal bodies. At the same time, the function of this protein in the repair of DNA double-strand breaks is enhanced. Following irradiation, cells overexpressing WRAP53ß exhibit more rapid clearance of phospho-histone H2AX (γH2AX), and more efficient homologous recombination and non-homologous end-joining, in association with fewer DNA breaks. Moreover, in these cells the ubiquitylation of damaged chromatin, which is known to facilitate the recruitment of repair factors and subsequent repair, is elevated. Knockdown of the ubiquitin ligase involved, ring-finger protein 8 (RNF8), which is recruited to DNA breaks by WRAP53ß, attenuated this effect, suggesting that overexpression of WRAP53ß leads to more rapid repair, as well as improved cell survival, by enhancing RNF8-mediated ubiquitylation at DNA breaks. Our present findings indicate that WRAP53ß and RNF8 are rate-limiting factors in the repair of DNA double-strand breaks and raise the possibility that upregulation of WRAP53ß may contribute to genomic stability in and survival of cancer cells.


Assuntos
Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Osteoblastos/metabolismo , Reparo de DNA por Recombinação , Telomerase/genética , Linhagem Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Corpos Enovelados/genética , Corpos Enovelados/metabolismo , DNA/química , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Telomerase/metabolismo , Telômero/química , Telômero/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
12.
Oncogene ; 35(6): 761-70, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25961912

RESUMO

The transcriptional cofactor PC4 is an ancient single-strand DNA (ssDNA)-binding protein that has a homologue in bacteriophage T5 where it is likely the elusive replicative ssDNA-binding protein. We hypothesize that human PC4 has retained functions in ssDNA binding to stabilize replication forks and prevent genome instability in mammalian cells. Here we demonstrate that PC4 is recruited to hydroxyurea (HU)-stalled replication forks, which is dependent on active transcription and its ssDNA-binding ability. Interestingly, we demonstrate that ssDNA binding by PC4 is critical to suppress spontaneous DNA damage and promote cellular survival. PC4 accumulation co-localizes with replication protein A (RPA) at stalled forks and is increased upon RPA depletion, demonstrating compensatory functions in ssDNA binding. Depletion of PC4 not only results in defective resolution of HU-induced DNA damage but also significantly reduces homologous recombination repair efficiency. Altogether, our results indicate that PC4 has similar functions to RPA in binding ssDNA to promote genome stability, especially at sites of replication-transcription collisions.


Assuntos
Dano ao DNA , Reparo do DNA/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/fisiologia , Instabilidade Genômica/genética , Fatores de Transcrição/fisiologia , Células Cultivadas , Quebras de DNA de Cadeia Dupla , Quebras de DNA de Cadeia Simples , Dano ao DNA/genética , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Humanos , Mutação , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
14.
Oncogene ; 32(32): 3744-53, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22945645

RESUMO

It has become increasingly clear that oncogenes not only provide aberrant growth signals to cells but also cause DNA damage at replication forks (replication stress), which activate the ataxia telangiectasia mutated (ATM)/p53-dependent tumor barrier. Here we studied underlying mechanisms of oncogene-induced replication stress in cells overexpressing the oncogene Cyclin E. Cyclin E overexpression is associated with increased firing of replication origins, impaired replication fork progression and DNA damage that activates RAD51-mediated recombination. By inhibiting replication initiation factors, we show that Cyclin E-induced replication slowing and DNA damage is a consequence of excessive origin firing. A significant amount of Cyclin E-induced replication slowing is due to interference between replication and transcription, which also underlies the activation of homologous recombination. Our data suggest that Cyclin E-induced replication stress is caused by deregulation of replication initiation and increased interference between replication and transcription, which results in impaired replication fork progression and DNA damage triggering the tumor barrier or cancer-promoting mutations.


Assuntos
Ciclina E/fisiologia , Replicação do DNA , Transcrição Gênica , Linhagem Celular Tumoral , Dano ao DNA , Recombinação Homóloga , Humanos , Oncogenes , Rad51 Recombinase/fisiologia
15.
J Appl Microbiol ; 105(1): 290-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18284484

RESUMO

AIMS: To investigate if the site-specific tyrosine integrase (Int) from phage P2 has features that would make it interesting for use of gene transfer into eukaryotic cells. These include the possibility of promoting recombination with a nonphage sequence, abolishing the requirement for the bacterial DNA-binding and -bending protein integration host factor (IHF), and localization to the nucleus of eukaryotic cells. METHODS AND RESULTS: We show that the Int protein catalyzes site-specific recombination using a human sequence in Escherichia coli and in vitro although not as efficiently as with the wild-type bacterial sequence, and that insertion of high mobility group recognition boxes in the phage attachment site substrate abolish the requirement of IHF and allows efficient recombination in vitro in a eukaryotic cell extract. Furthermore, we show by fluorescence that the Int protein contains a functional intrinsic nuclear localization signal, localizing it to the nucleus in both HeLa and 293 cells. CONCLUSIONS: We conclude that P2 Int may be a potential tool for site-specific integration of genes into the human chromosome. SIGNIFICANCE AND IMPACT OF THE STUDY: The study implies the possibility of using multiple prokaryotic Int proteins with different specific integration sites in human cells for future gene therapy programmes.


Assuntos
Sítios de Ligação Microbiológicos , Bacteriófago P2/enzimologia , Células Eucarióticas/virologia , Integrases/genética , Recombinação Genética , Animais , Bacteriófago P2/genética , Catálise , Núcleo Celular/virologia , DNA Viral/genética , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/virologia , Células Eucarióticas/metabolismo , Engenharia Genética , Terapia Genética/métodos , Células HeLa , Humanos , Reação em Cadeia da Polimerase/métodos , Coelhos , Transfecção/métodos , Integração Viral
16.
Biochem Soc Trans ; 35(Pt 5): 1352-4, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956349

RESUMO

DNA lesions resulting from impaired progression of replication forks are implicated in genetic instability and tumorigenesis. Because the cellular response to these lesions poses an important tumorigenesis barrier, the responsible signalling and repair pathways are often mutated or inactive in tumours. Here, we discuss how such deficiencies can in turn be exploited for cancer therapy.


Assuntos
Transformação Celular Neoplásica , Replicação do DNA , Neoplasias/terapia , Reparo do DNA , Humanos
17.
Cytogenet Genome Res ; 116(1-2): 38-45, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17268176

RESUMO

The RAD51C (RAD51L2) protein is one out of five RAD51 paralogs and forms a complex that includes either XRCC2 or XRCC3. Both of these complexes may have important functions in homologous recombination (HR). Here, we confirm that the frequency of DNA double-strand break (DSB)-induced HR is reduced in the RAD51C deficient cell line CL-V4B, in agreement with a role for RAD51C in HR. We report that mitotic RAD51C deficient CL-V4B cells also have an increased number of centrosomes in mitosis resulting in aberrant mitotic spindles. These data suggest that the RAD51C protein is important in maintaining correct centrosome numbers and that the complexes including RAD51C and XRCC2 or XRCC3 may be of importance in maintaining correct centrosome numbers in mitosis. Increased centrosome numbers following a RAD51C defect indicates that this protein might be important in preventing aneuploidy, suggesting that it could be a potential tumour suppressor in mammals.


Assuntos
Centrossomo/ultraestrutura , Proteínas de Ligação a DNA/fisiologia , Mitose , Recombinação Genética , Fuso Acromático , Animais , Células CHO , Linhagem Celular , Centrossomo/metabolismo , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/química , Genes Reporter , Genótipo , Microscopia de Fluorescência
18.
Biochem Soc Trans ; 32(Pt 6): 959-61, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15506935

RESUMO

PARP [poly(ADP-ribose) polymerase] activity is up-regulated by binding to DNA strand breaks and its association with DNA repair is well documented. Many anticancer therapies work by inducing breaks in DNA, if unrepaired these can lead to cell death. As PARP promotes DNA repair there is a strong rational to suggest that its inhibition may increase the efficiency of certain cytotoxic treatments. This review discusses the advances made in PARP inhibitor design and the mechanism by which they enhance anti-tumour therapies.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases , Azulenos , Benzodiazepinas/farmacologia , Replicação do DNA/efeitos dos fármacos , Desenho de Fármacos , Humanos
19.
Mutat Res ; 493(1-2): 55-66, 2001 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-11516715

RESUMO

Extremely low-frequency (ELF) magnetic fields have previously been shown to affect conformation of chromatin, cell proliferation, and calcium metabolism. Possible mutagenic and carcinogenic effects of ELF have also been discussed and tested. In this study, intrachromosomal recombination in the hprt gene after exposure to ELF magnetic field was investigated using the SPD8 recombination assay. SPD8 cells, derived from V79 Chinese hamster cells were exposed to ELF at a specific combination of static and ELF magnetic fields, that has been proven to have effects on chromatin conformation in several cell types. The genotoxic agent camptothecin (CPT) was used either as a positive control or simultaneously with ELF. We also analysed the effect of ELF and CPT on chromatin conformation with the anomalous viscosity time dependence (AVTD) technique, cell growth kinetics, and cell survival with clonogenic assay. DNA fragmentation was analysed by pulsed field gel electrophoresis (PFGE). ELF did not induce recombination alone, neither did ELF modify the recombinogenic effect of CPT. Although, there was no effect on cell survival in response to ELF exposure, inhibition of cell growth was observed. On the other hand, ELF exposure partly counteracted the growth inhibition seen with CPT. The data suggest that ELF exposure may stimulate or inhibit cell growth depending on the state of the cells. Although, ELF did not induce recombination, a weak but statistically significant DNA fragmentation comparable with CPT-induced fragmentation was observed with PFGE 48h after exposure to ELF.


Assuntos
Divisão Celular , Magnetismo/efeitos adversos , Recombinação Genética , Animais , Camptotecina/toxicidade , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Fragmentação do DNA/efeitos dos fármacos , Mutagênicos/toxicidade , Mutação , Recombinação Genética/efeitos dos fármacos
20.
Altern Lab Anim ; 29(3): 233-41, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11387020

RESUMO

A high throughput assay (the DRAG test) is described, which could be a useful tool for the detection of repairable DNA adducts, and which is based on the inhibition of the growth of DNA repair-deficient Chinese hamster ovary (CHO) cells. The cytotoxicity of a test substance towards DNA repair-deficient CHO cell lines is compared with the corresponding cytotoxicity in the parental wild-type CHO cell line (AA8). A more pronounced toxicity toward a DNA repair-deficient cell line is interpreted as being the consequence of its inability to repair the DNA adduct induced by the compound. (+)-7beta,8alpha-Dihydroxy-9alpha,10alpha-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene, camptothecin, ethyl methanesulphonate and mitomycin C were used as reference substances, and the overall results indicate that the DRAG test could be useful in the screening of compounds for the production of repairable DNA adducts. The main advantages with the DRAG test are that it provides a relevant endpoint, it is rapid, it requires small amounts of the test item, and it permits a large number of compounds to be tested.


Assuntos
Adutos de DNA/análise , Reparo do DNA , Testes de Mutagenicidade/métodos , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Animais , Células CHO , Camptotecina/toxicidade , Carcinógenos/toxicidade , Divisão Celular/efeitos dos fármacos , Cricetinae , Dano ao DNA , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Mesilatos/toxicidade , Mitomicina/toxicidade , Mutagênicos/toxicidade , Padrões de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...