Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 8(67): 110983-110993, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29340031

RESUMO

The combination of radiotherapy and pharmacological inhibition of cellular signal transduction pathways offers promising strategies for enhanced cancer cell inactivation. However, the molecular effects of kinase inhibitors especially on DNA damage detection and repair after X-irradiation have to be understood to facilitate the development of efficient and personalized treatment regimens. Therefore, we applied differential proteomics for analyzing inhibitor-induced changes in either chromatin-bound or phosphorylated nuclear proteins. The effect of the multi kinase inhibitor sorafenib on DNA repair, chromatin binding and phosphorylation of nuclear proteins was analyzed in UT-SCC 42B head and neck cancer cells using metabolic labeling based differential proteomics (SILAC). Sorafenib significantly inhibited DNA repair but failed to significantly affect chromatin interactions of 90 quantified proteins. In contrast, analyzing nuclear phospho-proteins following sorafenib treatment, we detected quantitative changes in 9 out of 59 proteins, including DNA-repair proteins. In conclusion, the analysis of nuclear phospho-proteins by differential proteomics is an effective tool for determining the molecular effects of kinase inhibitors on X-irradiated cells. Analyzing chromatin binding might be less promising.

2.
Oncotarget ; 7(29): 45122-45133, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27281611

RESUMO

The increase in cellular radiosensitivity by EGF receptor (EGFR) inhibition has been shown to be attributable to the induction of a G1-arrest in p53-proficient cells. Because EGFR targeting in combination with radiotherapy is used to treat head and neck squamous cell carcinomas (HNSCC) which are predominantly p53 mutated, we tested the effects of EGFR targeting on cellular radiosensitivity, proliferation, apoptosis, DNA repair and cell cycle control using a large panel of HNSCC cell lines. In these experiments EGFR targeting inhibited signal transduction, blocked proliferation and induced radiosensitization but only in some cell lines and only under normal (pre-plating) conditions. This sensitization was not associated with impaired DNA repair (53BP1 foci) or induction of apoptosis. However, it was associated with the induction of a lasting G2-arrest. Both, the radiosensitization and the G2-arrest were abrogated if the cells were re-stimulated (delayed plating) with actually no radiosensitization being detectable in any of the 14 tested cell lines. Therefore we conclude that EGFR targeting can induce a reversible G2 arrest in p53 deficient HNSCC cells, which does not consequently result in a robust cellular radiosensitization. Together with recent animal and clinical studies our data indicate that EGFR inhibition is no effective strategy to increase the radiosensitivity of HNSCC cells.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Receptores ErbB/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/patologia , Tolerância a Radiação/efeitos dos fármacos , Carcinoma de Células Escamosas/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cetuximab/farmacologia , Cloridrato de Erlotinib/farmacologia , Neoplasias de Cabeça e Pescoço/genética , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Proteína Supressora de Tumor p53/genética
3.
Radiother Oncol ; 116(3): 423-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26422459

RESUMO

BACKGROUND: EGFR inhibition blocks DNA double strand break (DSB) repair but the detailed mechanisms are still unclear. We asked whether EGFR inhibition blocks DSB repair by reducing the X-ray-induced phosphorylation of repair proteins using a phosphoproteomic approach. MATERIALS AND METHODS: Using UT-SCC5 and SAS head and neck cancer cells we established a differential phosphoproteomic approach for quantitative analysis of DNA repair proteins by stable isotope labeling with amino acids. Nuclear phosphoproteins were isolated and analyzed by liquid chromatography/tandem mass spectrometry. Erlotinib, PD98059 and olaparib were used to inhibit EGFR, MEK1/2 and PARP1, respectively. PARP1 was knocked down by siRNA. DSB repair was measured by quantifying residual 53BP1 foci. RESULTS: Over 150 nuclear phosphoproteins were quantified after irradiation, including 24 DNA repair proteins. Two of these, including PARP1, were consistently reduced in both cell lines upon erlotinib treatment. PARP1 inhibition or knock-down and EGFR inhibition resulted in an analog number of residual foci which was not further increased by combination of both strategies. MEK1/2 inhibition with or without blockage of EGFR or PARP1 caused similar effects. CONCLUSION: We have established a powerful, quantitative phosphoproteomic approach to investigate regulatory mechanisms in DSB repair, dependent on protein phosphorylation after irradiation. Using this approach we have identified PARP1 as a mediator of EGFR/MEK-dependent regulation of DSB repair.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA/fisiologia , Receptores ErbB/fisiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Proteômica , Carcinoma de Células Escamosas/genética , DNA/genética , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib/farmacologia , Flavonoides/farmacologia , Neoplasias de Cabeça e Pescoço/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosforilação/efeitos da radiação , Ftalazinas/farmacologia , Piperazinas/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Células Tumorais Cultivadas , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
4.
Radiother Oncol ; 101(1): 147-51, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21665306

RESUMO

PURPOSE: The purpose of this study was to examine whether the epidermal growth factor receptor (EGFR) may be used as a general target to modulate DNA double strand break (DSB) repair in tumor cells. MATERIAL AND METHODS: Experiments were performed with human tumor cell lines A549, H1299 and HeLa and primate cell line CV1. EGF, ARG and TGFα were used for EGFR activation, cetuximab or erlotinib for inhibition. Overall DSB repair was assessed by γH2AX/53BP1 co-immunostaining and non-homologous end-joining (NHEJ) and homologous recombination (HR) by using NHEJ and HR reporter cells; cell cycle distribution was determined by flow cytometry and protein expression by Western blot. RESULTS: EGFR activation was found to stimulate overall DSB repair as well as NHEJ regardless of the ligand used. This stimulation was abolished when EGFR signaling was blocked. This regulation was found for all cell lines tested, irrespective of their p53 or K-Ras status. Stimulation and inhibition of EGFR were also found to affect HR. CONCLUSIONS: Regulation of DSB repair by EGFR involves both the NHEJ and HR pathway, and appears to occur in most tumor cell lines regardless of p53 and K-Ras mutation status.


Assuntos
Carcinoma Broncogênico/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Receptores ErbB/metabolismo , Recombinação Homóloga/genética , Neoplasias do Colo do Útero/genética , Animais , Western Blotting , Carcinoma Broncogênico/radioterapia , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Feminino , Fibroblastos/efeitos da radiação , Imunofluorescência , Genes p53/genética , Genes p53/efeitos da radiação , Genes ras/genética , Haplorrinos , Humanos , Radiação Ionizante , Células Tumorais Cultivadas/efeitos da radiação , Neoplasias do Colo do Útero/radioterapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...