Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Med ; 29(1): 15, 2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36717782

RESUMO

BACKGROUND: Osteosarcoma is a malignant bone tumor that usually affects adolescents aged 15-19 y. The DNA damage response (DDR) is significantly enhanced in osteosarcoma, impairing the effect of systemic chemotherapy. Targeting the DDR process was considered a feasible strategy benefitting osteosarcoma patients. However, the clinical application of DDR inhibitors is not impressive because of their side effects. Chinese herbal medicines with high anti-tumor effects and low toxicity in the human body have gradually gained attention. 2-Hydroxy-3-methylanthraquinone (HMA), a Chinese medicine monomer found in the extract of Oldenlandia diffusa, exerts significant inhibitory effects on various tumors. However, its anti-osteosarcoma effects and defined molecular mechanisms have not been reported. METHODS: After HMA treatment, the proliferation and metastasis capacity of osteosarcoma cells was detected by CCK-8, colony formation, transwell assays and Annexin V-fluorescein isothiocyanate/propidium iodide staining. RNA-sequence, plasmid infection, RNA interference, Western blotting and immunofluorescence assay were used to investigate the molecular mechanism and effects of HMA inhibiting osteosarcoma. Rescue assay and CHIP assay was used to further verified the relationship between MYC, CHK1 and RAD51. RESULTS: HMA regulate MYC to inhibit osteosarcoma proliferation and DNA damage repair through PI3K/AKT signaling pathway. The results of RNA-seq, IHC, Western boltting etc. showed relationship between MYC, CHK1 and RAD51. Rescue assay and CHIP assay further verified HMA can impair homologous recombination repair through the MYC-CHK1-RAD51 pathway. CONCLUSION: HMA significantly inhibits osteosarcoma proliferation and homologous recombination repair through the MYC-CHK1-RAD51 pathway, which is mediated by the PI3K-AKT signaling pathway. This study investigated the exact mechanism of the anti-osteosarcoma effect of HMA and provided a potential feasible strategy for the clinical treatment of human osteosarcoma.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Adolescente , Reparo de DNA por Recombinação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Rad51 Recombinase/farmacologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células
2.
Int J Mol Sci ; 22(18)2021 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-34575923

RESUMO

Molecules involved in DNA damage response (DDR) are often overexpressed in cancer cells, resulting in poor responses to chemotherapy and radiotherapy. Although treatment efficacy can be improved with the concomitant use of DNA repair inhibitors, the accompanying side effects can compromise the quality of life of patients. Therefore, in this study, we identified a natural compound that could inhibit DDR, using the single-strand annealing yeast-cell analysis system, and explored its mechanisms of action and potential as a chemotherapy adjuvant in hepatocellular carcinoma (HCC) cell lines using comet assay, flow cytometry, Western blotting, immunofluorescence staining, and functional analyses. We developed a mouse model to verify the in vitro findings. We found that hydroxygenkwanin (HGK) inhibited the expression of RAD51 and progression of homologous recombination, thereby suppressing the ability of the HCC cell lines to repair DNA damage and enhancing their sensitivity to doxorubicin. HGK inhibited the phosphorylation of DNA damage checkpoint proteins, leading to apoptosis in the HCC cell lines. In the mouse xenograft model, HGK enhanced the sensitivity of liver cancer cells to doxorubicin without any physiological toxicity. Thus, HGK can inhibit DDR in liver cancer cells and mouse models, making it suitable for use as a chemotherapy adjuvant.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Dano ao DNA/efeitos dos fármacos , Flavonoides/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Reparo do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Medicamentos de Ervas Chinesas , Regulação da Expressão Gênica , Recombinação Homóloga/efeitos dos fármacos , Humanos , Camundongos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Leveduras/efeitos dos fármacos , Leveduras/genética , Leveduras/metabolismo
3.
Plant Cell ; 33(8): 2869-2882, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34009315

RESUMO

Meiosis is a fundamental process for sexual reproduction in most eukaryotes and the evolutionarily conserved recombinases RADiation sensitive51 (RAD51) and Disrupted Meiotic cDNA1 (DMC1) are essential for meiosis and thus fertility. The mitotic function of RAD51 is clear, but the meiotic function of RAD51 remains largely unknown. Here we show that RAD51 functions as an interacting protein to restrain the Structural Maintenance of Chromosomes5/6 (SMC5/6) complex from inhibiting DMC1. We unexpectedly found that loss of the SMC5/6 partially suppresses the rad51 knockout mutant in terms of sterility, pollen inviability, and meiotic chromosome fragmentation in a DMC1-dependent manner in Arabidopsis thaliana. Biochemical and cytological studies revealed that the DMC1 localization in meiotic chromosomes is inhibited by the SMC5/6 complex, which is attenuated by RAD51 through physical interactions. This study not only identified the long-sought-after function of RAD51 in meiosis but also discovered the inhibition of SMC5/6 on DMC1 as a control mechanism during meiotic recombination.


Assuntos
Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Rad51 Recombinase/genética , Recombinases Rec A/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Pareamento Cromossômico , Cromossomos de Plantas , Regulação da Expressão Gênica de Plantas , Mutação com Perda de Função , Meiose , Complexos Multiproteicos/metabolismo , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Infertilidade das Plantas/genética , Pólen/genética , Rad51 Recombinase/metabolismo , Recombinases Rec A/genética
4.
Nat Commun ; 11(1): 2950, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32528002

RESUMO

During homologous recombination, Rad51 forms a nucleoprotein filament on single-stranded DNA to promote DNA strand exchange. This filament binds to double-stranded DNA (dsDNA), searches for homology, and promotes transfer of the complementary strand, producing a new heteroduplex. Strand exchange proceeds via two distinct three-strand intermediates, C1 and C2. C1 contains the intact donor dsDNA whereas C2 contains newly formed heteroduplex DNA. Here, we show that the conserved DNA binding motifs, loop 1 (L1) and loop 2 (L2) in site I of Rad51, play distinct roles in this process. L1 is involved in formation of the C1 complex whereas L2 mediates the C1-C2 transition, producing the heteroduplex. Another DNA binding motif, site II, serves as the DNA entry position for initial Rad51 filament formation, as well as for donor dsDNA incorporation. Our study provides a comprehensive molecular model for the catalytic process of strand exchange mediated by eukaryotic RecA-family recombinases.


Assuntos
DNA/metabolismo , Rad51 Recombinase/química , Rad51 Recombinase/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação/genética , DNA/genética , Dano ao DNA/genética , Dano ao DNA/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , DNA de Cadeia Simples/genética , Recombinação Homóloga/genética , Recombinação Homóloga/fisiologia , Humanos , Mutação/genética , Ácidos Nucleicos Heteroduplexes/genética , Ácidos Nucleicos Heteroduplexes/metabolismo , Estrutura Secundária de Proteína , Rad51 Recombinase/genética , Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética
5.
PLoS One ; 14(5): e0216744, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31091255

RESUMO

OBJECTIVE: Aims were to investigate sensitivity of various human and canine cancer cell lines to hyperthermia and the influence of particular treatment conditions, and to analyze the DNA-damage response and mode of cell death in cell line radiosensitized by hyperthermia. Additionally, we were interested in the involvement of HSP70 in radiosensitization. METHODS: Radiosensitization by hyperthermia was determined in a panel of human and canine cancer cell lines using clonogenic cell survival assay, as well as levels of heat shock proteins (HSPs) using immunoblotting. The influence of the hyperthermia-radiotherapy time gap, different temperatures and the order of treatments on clonogenicity of hyperthermia-sensitive A549 cells was investigated. Additionally, DNA damage and cell death were assessed by Comet assay and an apoptosis/necrosis assay. Further we induced transient knockdown in A549 cells to test HSP70's involvement in radiosensitization. RESULTS: Out of eight cell lines tested, only two (A549 and Abrams) showed significant decrease in clonogenic cell survival when pre-treated with hyperthermia at 42°C. Strong induction of HSP70 upon thermoradiotherapy (HT-RT) treatment was found in all cell lines. Transient knockdown of HSP70 in A549 cells did not result in decrease of clonogenic cell survival in response to HT-RT. CONCLUSION: Tumor cell-type, temperature and order of treatment play an important role in radiosensitization by hyperthermia. However, hyperthermia has limited potency to radiosensitize canine cancer cells grown in a 2D cell culture setting presented here. DNA damage and apoptosis/necrosis did not increase upon combined treatment and cytosolic levels of HSP70 appear not to play critical role in the radiosensitization of A549 cells.


Assuntos
Hipertermia Induzida/métodos , Neoplasias/terapia , Tolerância a Radiação , Células A549 , Animais , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Dano ao DNA , Cães , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Rad51 Recombinase/metabolismo , Ensaio Tumoral de Célula-Tronco
6.
Sci Rep ; 9(1): 3842, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30846712

RESUMO

Green tea polyphenols may protect cells from UV damage through antioxidant activities and by stimulating the removal of damaged or cross-linked DNA. Recently, DNA repair pathways have been predicted as possible targets of epigallocatechin gallate (EGCG)-initiated signaling. However, whether and how green tea polyphenols can promote nucleotide excision repair and homologous recombination in diverse organisms requires further investigation. In this report, we used the budding yeast, Saccharomyces cerevisiae, as a model to investigate the effects of green tea extract on DNA repair pathways. We first showed that green tea extract increased the survival rate and decreased the frequency of mutations in yeast exposed to UVB-irradiation. Furthermore, green tea extract increased the expression of homologous recombination genes, RFA1, RAD51 and RAD52, and nucleotide excision repair genes, RAD4 and RAD14. Importantly, we further used a specific strand invasion assay to show that green tea extract promotes homologous recombination at double-strand breaks. Thus, green tea extract acts to preserve genome stability by activating DNA repair pathways in yeast. Because homologous recombination repair is highly conserved in yeast and humans, this study demonstrates yeast may be a useful platform for future research to investigate the underlying mechanisms of the bioactive compounds in DNA repair.


Assuntos
Reparo do DNA/efeitos dos fármacos , Extratos Vegetais/farmacologia , Saccharomyces cerevisiae/efeitos dos fármacos , Chá , DNA Fúngico/efeitos dos fármacos , DNA Fúngico/efeitos da radiação , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteína de Replicação A/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Raios Ultravioleta/efeitos adversos
7.
Cell Death Dis ; 9(10): 1025, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30287840

RESUMO

Procaspase-3-activating compound 1 (PAC-1) induces procaspase-3 activation via zinc chelation. However, whether PAC-1 employs other mechanisms remains unknown. Here we systematically screened for potent PAC-1 targets using 29 enhanced green fluorescent protein-labeled reporter cell lines and identified hypoxia-inducible factor 1α (HIF1α) and RAD51 pathways as PAC-1 targets. These results were verified in HepG2 cells and two other cancer cell lines. Mechanistically, PAC-1 specifically blocked HIF1α hydroxylation and upregulated HIF1α target genes. In addition, DNA damage, G1/S cell cycle arrest, and the inhibition of DNA synthesis were induced following PAC-1 administration. Interestingly, by using ferrozine-iron sequestration and iron titration assays, we uncovered the iron sequestering capacity of PAC-1. Additionally, the expression levels of iron shortage-related genes were also increased in PAC-1-treated cells, and iron (II) supplementation reversed all of the observed cellular responses. Thus, our results indicate that PAC-1 induces HIF1α stabilization and DNA damage by sequestering ferrous iron.


Assuntos
Caspase 3/metabolismo , Dano ao DNA/efeitos dos fármacos , Ferrozina/farmacologia , Hidrazonas/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/tratamento farmacológico , Ferro/farmacologia , Piperazinas/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Hipóxia/metabolismo , Rad51 Recombinase/metabolismo , Regulação para Cima/efeitos dos fármacos , Zinco/metabolismo
8.
DNA Repair (Amst) ; 60: 89-101, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29112893

RESUMO

Investigation of natural products is an attractive strategy to identify novel compounds for cancer prevention and treatment. Numerous studies have shown the efficacy and safety of natural products, and they have been widely used as alternative treatments for a wide range of illnesses, including cancers. However, it remains unknown whether natural products affect homologous recombination (HR)-mediated DNA repair and whether these compounds can be used as sensitizers with minimal toxicity to improve patients' responses to radiation therapy, a mainstay of treatment for many human cancers. In this study, in order to systematically identify natural products with an inhibitory effect on HR repair, we developed a high-throughput image-based HR repair screening assay and screened a chemical library containing natural products. Among the most interesting of the candidate compounds identified from the screen was ß-thujaplicin, a bioactive compound isolated from the heart wood of plants in the Cupressaceae family, can significantly inhibit HR repair. We further demonstrated that ß-thujaplicin inhibits HR repair by reducing the recruitment of a key HR repair protein, Rad51, to DNA double-strand breaks. More importantly, our results showed that ß-thujaplicin can radiosensitize cancer cells. Additionally, ß-thujaplicin sensitizes cancer cells to PARP inhibitor in different cancer cell lines. Collectively, our findings for the first time identify natural compound ß-thujaplicin, which has a good biosafety profile, as a novel HR repair inhibitor with great potential to be translated into clinical applications as a sensitizer to DNA-damage-inducing treatment such as radiation and PARP inhibitor. In addition, our study provides proof of the principle that our robust high-throughput functional HR repair assay can be used for a large-scale screening system to identify novel natural products that regulate DNA repair and cellular responses to DNA damage-inducing treatments such as radiation therapy.


Assuntos
Monoterpenos/uso terapêutico , Neoplasias/tratamento farmacológico , Rad51 Recombinase/efeitos dos fármacos , Radiossensibilizantes/uso terapêutico , Reparo de DNA por Recombinação/efeitos dos fármacos , Tropolona/análogos & derivados , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Cupressaceae/química , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Quimioterapia Combinada , Feminino , Ensaios de Triagem em Larga Escala , Humanos , Monoterpenos/farmacologia , Monoterpenos/toxicidade , Neoplasias/enzimologia , Neoplasias/genética , Extratos Vegetais , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Rad51 Recombinase/metabolismo , Radiossensibilizantes/farmacologia , Radiossensibilizantes/toxicidade , Tropolona/farmacologia , Tropolona/uso terapêutico , Tropolona/toxicidade
9.
Oncotarget ; 8(27): 44593-44604, 2017 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-28574821

RESUMO

Hyperthermia has a number of biological effects that sensitize tumors to radiotherapy in the range between 40-44 °C. One of these effects is heat-induced degradation of BRCA2 that in turn causes reduced RAD51 focus formation, which results in an attenuation of DNA repair through homologous recombination. Prompted by this molecular insight into how hyperthermia attenuates homologous recombination, we now quantitatively explore time and temperature dynamics of hyperthermia on BRCA2 levels and RAD51 focus formation in cell culture models, and link this to their clonogenic survival capacity after irradiation (0-6 Gy). For treatment temperatures above 41 °C, we found a decrease in cell survival, an increase in sensitization towards irradiation, a decrease of BRCA2 protein levels, and altered RAD51 focus formation. When the temperatures exceeded 43 °C, we found that hyperthermia alone killed more cells directly, and that processes other than homologous recombination were affected by the heat. This study demonstrates that optimal inhibition of HR is achieved by subjecting cells to hyperthermia at 41-43 °C for 30 to 60 minutes. Our data provides a guideline for the clinical application of novel combination treatments that could exploit hyperthermia's attenuation of homologous recombination, such as the combination of hyperthermia with PARP-inhibitors for non-BRCA mutations carriers.


Assuntos
Dano ao DNA , Reparo do DNA , Recombinação Homóloga , Hipertermia Induzida , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Humanos , Transporte Proteico , Proteólise , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Tolerância a Radiação/genética , Temperatura , Fatores de Tempo
10.
Oncotarget ; 7(38): 62585-62597, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27566579

RESUMO

(-)-Guaiol, generally known as an antibacterial compound, has been found in many medicinal plants. Its roles in tumor suppression are still under investigation. In the study, we mainly focused on exploring its applications in dealing with non-small cell lung cancer (NSCLC) and the underlying mechanisms. Here, we show that (-)-Guaiol significantly inhibits cell growth of NSCLC cells both in vitro and in vivo. Further high throughput analysis reveals that RAD51, a pivotal factor in homologous recombination repair, is a potential target for it. The following mechanism studies show that (-)-Guaiol is involved in cell autophagy to regulate the expression of RAD51, leading to double-strand breaks triggered cell apoptosis. Moreover, targeting RAD51, which is highly overexpressed in the lung adenocarcinoma tissues, can significantly increase the chemosensitivity of NSCLC cells to (-)-Guaiol both in vitro and in vivo. All in all, our studies provide an attractive insight in applying (-)-Guaiol into NSCLC treatments and further suggest that knockdown of oncogenic RAD51 will greatly enhance the chemosensitivity of patients with NSCLC.


Assuntos
Adenocarcinoma/metabolismo , Apoptose , Autofagia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Rad51 Recombinase/metabolismo , Sesquiterpenos/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma de Pulmão , Animais , Antineoplásicos/química , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oncogenes , Recombinação Genética , Sesquiterpenos de Guaiano
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 940-4, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-27363277

RESUMO

Objective To investigate the effect of evodiamine on the radiosensitivity of esophageal squamous cell cancer Eca-109 cells. Methods Eca-109 cells were treated with various concentrations of evodiamine [(10, 20, 40, 60, 80, 100, 120) µg/mL], and then cell proliferation was examined by MTT assay. After the optimal evodiamine concentration was determined, the cells were divided into radiation group (0, 2, 4, 6, 8 Gy X-ray radiation) and radiation combined with evodiamine group (80 µg/mL evodiamine and 0, 2, 4, 6, 8 Gy X-ray radiation) .The radiosensitivity of Eca-109 cells was detected using colony formation assay. Flow cytometry was used to determine cell cycle of Eca-109 cells. The protein expressions of Ku70, Ku80, DNA-PKcs and Rad51 were examined by Western blotting. Results MTT assay showed that evodiamine decreased the proliferation of Eca-109 cells in a concentration-dependent manner. The inhibition reached the maximal level at 80 µg/mL. Compared with radiotherapy alone, the combination of 80 µg/mL evodiamine and radiotherapy improved survival curve and decreased the values of D0 and Dq. Sensitizer enhancement ratio was 1.86±0.06. Furthermore, cell cycle analysis revealed that evodiamine suppressed radiotherapy-induced the G2/M arrest. Additionally, evodiamine treatment also significantly inhibited radiotherapy-induced increase in Ku70, Ku80, DNA-PKcs and Rad51 expressions. Conclusion Evodiamine enhances radiosensitivity of Eca-109 cells during radiotherapy. The effect may be associated with the inhibition of G2/M arrest and the attenuation of Ku70, Ku80, DNA-PKcs and Rad51 expressions.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Quinazolinas/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Proteína Quinase Ativada por DNA/metabolismo , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Humanos , Autoantígeno Ku/metabolismo , Proteínas Nucleares/metabolismo , Extratos Vegetais/farmacologia , Rad51 Recombinase/metabolismo , Raios X
12.
Int J Cancer ; 139(2): 467-79, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26933761

RESUMO

The tetrahydroisoquinoline trabectedin is a marine compound with approved activity against human soft-tissue sarcoma. It exerts antiproliferative activity mainly by specific binding to the DNA and inducing DNA double-strand breaks (DSB). As homologous recombination repair (HRR)-deficient tumors are more susceptible to trabectedin, hyperthermia-mediated on-demand induction of HRR deficiency represents a novel and promising strategy to boost trabectedin treatment. For the first time, we demonstrate enhancement of trabectedin effectiveness in human sarcoma cell lines by heat and characterize cellular events and molecular mechanisms related to heat-induced effects. Hyperthermic temperatures (41.8 or 43°C) enhanced significantly trabectedin-related clonogenic cell death and G2/M cell cycle arrest followed by cell type-dependent induction of apoptosis or senescence. Heat combination increased accumulation of γH2AX foci as key marker of DSBs. Expression of BRCA2 protein, an integral protein of the HRR machinery, was significantly decreased by heat. Consequently, recruitment of downstream RAD51 to γH2AX-positive repair foci was almost abolished indicating relevant impairment of HRR by heat. Accordingly, enhancement of trabectedin effectiveness was significantly augmented in BRCA2-proficient cells by hyperthermia and alleviated in BRCA2 knockout or siRNA-transfected BRCA2 knockdown cells. In peripheral blood mononuclear cells isolated from sarcoma patients, increased numbers of nuclear γH2AX foci were detected after systemic treatment with trabectedin and hyperthermia of the tumor region. The findings establish BRCA2 degradation by heat as a key factor for a novel treatment strategy that allows targeted chemosensitization to trabectedin and other DNA damaging antitumor drugs by on-demand induction of HRR deficiency.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Proteína BRCA2/metabolismo , Dioxóis/farmacologia , Hipertermia Induzida , Reparo de DNA por Recombinação/efeitos dos fármacos , Reparo de DNA por Recombinação/efeitos da radiação , Tetra-Hidroisoquinolinas/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Histonas/metabolismo , Humanos , Modelos Biológicos , Ligação Proteica , Transporte Proteico , Proteólise/efeitos dos fármacos , Proteólise/efeitos da radiação , Rad51 Recombinase/metabolismo , Sarcoma/metabolismo , Sarcoma/patologia , Sarcoma/terapia , Trabectedina
13.
J Pineal Res ; 58(2): 189-97, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25623566

RESUMO

Radiation and adjuvant endocrine therapy are nowadays considered a standard treatment option after surgery in breast cancer. Melatonin exerts oncostatic actions on human breast cancer cells. In the current study, we investigated the effects of a combination of radiotherapy and melatonin on human breast cancer cells. Melatonin (1 mm, 10 µm and 1 nm) significantly inhibited the proliferation of MCF-7 cells. Radiation alone inhibited the MCF-7 cell proliferation in a dose-dependent manner. Pretreatment of breast cancer cells with melatonin 1 wk before radiation led to a significantly greater decrease of MCF-7 cell proliferation compared with radiation alone. Melatonin pretreatment before radiation also decreased G2 -M phase arrest compared with irradiation alone, with a higher percentage of cells in the G0 -G1 phase and a lower percentage of cells in S phase. Radiation alone diminished RAD51 and DNA-protein kinase (PKcs) mRNA expression, two main proteins involved in double-strand DNA break repair. Treatment with melatonin for 7 days before radiation led to a significantly greater decrease in RAD51 and DNA-PKcs mRNA expression compared with radiation alone. Our findings suggest that melatonin pretreatment before radiation sensitizes breast cancer cells to the ionizing effects of radiation by decreasing cell proliferation, inducing cell cycle arrest and downregulating proteins involved in double-strand DNA break repair. These findings may have implications for designing clinical trials using melatonin and radiotherapy.


Assuntos
Neoplasias da Mama/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Melatonina/farmacologia , Radiação Ionizante , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Quebras de DNA de Cadeia Dupla , Feminino , Humanos , Células MCF-7 , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
14.
J Biol Chem ; 289(33): 22614-22629, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24990942

RESUMO

Yin Yang 1 (YY1) is a transcription factor regulating proliferation and differentiation and is involved in cancer development. Oligomers of recombinant YY1 have been observed before, but their structure and DNA binding properties are not well understood. Here we find that YY1 assembles several homo-oligomeric species built from the association of a bell-shaped dimer, a process we characterized by electron microscopy. Moreover, we find that YY1 self-association also occurs in vivo using bimolecular fluorescence complementation. Unexpectedly, these oligomers recognize several DNA substrates without the consensus sequence for YY1 in vitro, and DNA binding is enhanced in the presence of RuvBL1-RuvBL2, two essential AAA+ ATPases. YY1 oligomers bind RuvBL1-RuvBL2 hetero-oligomeric complexes, but YY1 interacts preferentially with RuvBL1. Collectively, these findings suggest that YY1-RuvBL1-RuvBL2 complexes could contribute to functions beyond transcription, and we show that YY1 and the ATPase activity of RuvBL2 are required for RAD51 foci formation during homologous recombination.


Assuntos
Proteínas de Transporte/metabolismo , DNA Helicases/metabolismo , DNA/metabolismo , Complexos Multiproteicos/metabolismo , Multimerização Proteica/fisiologia , Fator de Transcrição YY1/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Proteínas de Transporte/genética , Linhagem Celular , DNA/genética , DNA Helicases/genética , Recombinação Homóloga/fisiologia , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/ultraestrutura , Ligação Proteica/fisiologia , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Transcrição Gênica/fisiologia , Fator de Transcrição YY1/genética
15.
PLoS One ; 8(8): e72431, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23951324

RESUMO

Two hallmark features of meiosis are i) the formation of crossovers (COs) between homologs and ii) the production of genetically-unique haploid spores that will fuse to restore the somatic ploidy level upon fertilization. In this study we analysed meiosis in haploid Arabidopsis thaliana plants and a range of haploid mutants to understand how meiosis progresses without a homolog. Extremely low chiasma frequency and very limited synapsis occurred in wild-type haploids. The resulting univalents segregated in two uneven groups at the first division, and sister chromatids segregated to opposite poles at the second division, leading to the production of unbalanced spores. DNA double-strand breaks that initiate meiotic recombination were formed, but in half the number compared to diploid meiosis. They were repaired in a RAD51- and REC8-dependent manner, but independently of DMC1, presumably using the sister chromatid as a template. Additionally, turning meiosis into mitosis (MiMe genotype) in haploids resulted in the production of balanced haploid gametes and restoration of fertility. The variability of the effect on meiosis of the absence of homologous chromosomes in different organisms is then discussed.


Assuntos
Arabidopsis/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Haploidia , Meiose/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Pareamento Cromossômico/genética , Troca Genética/genética , Diploide , Fertilidade/genética , Indóis/química , Mitose/genética , Proteína 1 Homóloga a MutL , Mutação , Pólen/genética , Pólen/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Recombinases Rec A/genética , Recombinases Rec A/metabolismo , Coloração e Rotulagem/métodos
16.
J Cell Physiol ; 228(7): 1473-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23254360

RESUMO

Hyperthermia has long been known as a radio-sensitizing agent that displays anti-tumor effects, and has been developed as a therapeutic application. The mechanisms of hyperthermia-induced radio-sensitization are highly associated with inhibition of DNA repair. Our investigations aimed to show how hyperthermia inactivate homologous recombination repair in the process of sensitizing cells to ionizing radiation by using a series of DNA repair deficient Chinese Hamster cells. Significant differences in cellular toxicity attributable to hyperthermia at and above 42.5°C were observed. In wild-type and non-homologous end joining repair mutants, cells in late S phase showed double the amount heat-induced radio-sensitization effects of G1-phase cells. Both radiation-induced DNA double strand breaks and chromatin damage resulting from hyperthermia exposure was measured to be approximately two times higher in G2-phase cells than G0/G1 cells. Additionally, G2-phase cells took approximately two times as long to repair DNA damage over time than G0/G1-phase cells. To supplement these findings, radiation-induced Rad51 foci formations at DNA double strand break sites were observed to gradually dissociate in response to the temperature and time of hyperthermia exposure. Dissociated Rad51 proteins subsequently re-formed foci at damage sites with time, and occurred in a trend also related to temperature and time of hyperthermia exposure. These findings suggest Rad51's dissociation and subsequent reformation at DNA double strand break sites in response to varying hyperthermia conditions plays an important role in hyperthermia-induced radio-sensitization.


Assuntos
Hipertermia Induzida , Tolerância a Radiação , Reparo de DNA por Recombinação , Animais , Células CHO , Ciclo Celular , Linhagem Celular , Sobrevivência Celular , Cricetinae , Cricetulus , Quebras de DNA de Cadeia Dupla , Histonas/metabolismo , Humanos , Mutação , Rad51 Recombinase/metabolismo , Reparo de DNA por Recombinação/genética , Temperatura , Fatores de Tempo
17.
J Exp Bot ; 63(14): 5323-35, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22859673

RESUMO

The RecA/RAD51 family of rice (Oryza sativa) consists of at least 13 members. However, the functions of most of these members are unknown. Here the functional characterization of one member of this family, RAD51C, is reported. Knockout (KO) of RAD51C resulted in both female and male sterility in rice. Transferring RAD51C to the RAD51C-KO line restored fertility. Cytological analyses showed that the sterility of RAD51C-KO plants was associated with abnormal early meiotic processes in both megasporocytes and pollen mother cells (PMCs). PMCs had an absence of normal pachytene chromosomes and had abnormal chromosome fragments. The RAD51C-KO line showed no obvious difference from wild-type plants in mitosis in the anther wall cells, which was consistent with the observation that the RAD51C-KO line did not have obviously abnormal morphology during vegetative development. However, the RAD51C-KO line was sensitive to different DNA-damaging agents. These results suggest that RAD51C is essential for reproductive development by regulating meiosis as well as for DNA damage repair in somatic cells.


Assuntos
Genes de Plantas , Oryza/citologia , Oryza/genética , Proteínas de Plantas/genética , Rad51 Recombinase/genética , Agrobacterium/genética , Cromossomos de Plantas/efeitos dos fármacos , Cromossomos de Plantas/metabolismo , Cromossomos de Plantas/efeitos da radiação , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , DNA Bacteriano/genética , Técnicas de Inativação de Genes , Teste de Complementação Genética , Células Germinativas Vegetais/efeitos dos fármacos , Células Germinativas Vegetais/crescimento & desenvolvimento , Células Germinativas Vegetais/efeitos da radiação , Meiose/efeitos dos fármacos , Meiose/efeitos da radiação , Mitose/efeitos dos fármacos , Mitose/efeitos da radiação , Dados de Sequência Molecular , Mutagênicos/farmacologia , Oryza/efeitos dos fármacos , Oryza/crescimento & desenvolvimento , Filogenia , Infertilidade das Plantas , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Pólen/efeitos dos fármacos , Pólen/crescimento & desenvolvimento , Pólen/efeitos da radiação , Rad51 Recombinase/metabolismo , Análise de Sequência de DNA
18.
Int J Mol Med ; 30(5): 1166-72, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22895634

RESUMO

Berberine, an isoquinoline derivative alkaloid, has recently been shown to have antitumor activity. The present study aimed to investigate the effects of the concomitant administration of berberine and radiation on breast cancer. The effects of berberine on the radiosensitivity of MCF-7 and MDA-MB-468 cells were evaluated by using cell clonogenic assays. Cells pre-treated with berberine or dimethyl sulfoxide (DMSO) for 24 h were irradiated using a Faxitron Cabinet X-ray System to deliver the indicated doses (0, 1, 2, 3 and 4 Gy). Changes in cell cycle distribution were determined by flow cytometry. γ-H2AX foci were detected by immunofluorescence staining. The levels of Ku70, Ku86 and RAD51 proteins were evaluated by western blot analysis. We observed that berberine increased the MCF-7 and MDA-MB-468 cell radiosensitivity with cell clonogenic assays. the radiation-induced G2/M cell cycle delay was reduced in the MCF-7 cells pre-teated with berberine. Berberine pre-treatment prolonged the persistence of DNA double-strand breaks in the MCF-7 cell line. In comparison with the control cells, the protein levels of RAD51 were decreased in the MCF-7 and MDA-MB-468 cells treated with berberine, and in the cells pre-treated with 15 µM berberine for 24 h, the level of RAD51 protein decreased significantly at the indicated time-points (0, 2, 6 and 24 h) following X-ray exposure. In conclusion, berberine sensitizes human breast cancer cells to ionizing radiation by inducing cell cycle arrest and the downregulation of the homologous recombination repair protein, RAD51. Berberine may be a promising radiosensitizer for the treatment of breast cancer.


Assuntos
Berberina/farmacologia , Radiossensibilizantes/farmacologia , Antígenos Nucleares/metabolismo , Neoplasias da Mama , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Histonas/metabolismo , Humanos , Autoantígeno Ku , Células MCF-7 , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
19.
Asian Pac J Cancer Prev ; 13(4): 1505-10, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22799356

RESUMO

To aim of this was to observe emodin-mediated cytotoxicity and its influence on Rad51 and ERCC1 expressionin non-small cell lung cancer (NSCLC). NSCLC cells were cultured in vitro with emodin at various concentrations (0, 25, 50, 75 and 100 µmol/L) for 48 h and the proliferation inhibition rate was determined by the MTT method. Then, NSCLC were treated with emodin (SK-MES-1 40 µmol/L, A549 70 µmol/L) or 20 µmol/L U0126 (an ERK inhibitor) for 48 h, or with various concentrations of emodin for 48 h and the protein and mRNA expressions of ERCC1 and Rad51 were determined by RT-PCR and Western blot assay, respectively. Emodin exerted a suppressive effect on the proliferation of NSCLC in a concentration dependent manner. Protein and mRNA expression of ERCC1 and Rad51 was also significantly decreased with the dose. Vacuolar degeneration was observed in A549 and SK-MES-1 cell lines after emodin treatment by transmission electron microscopy. Emodin may thus inhibited cell proliferation in NSCLC cells by downregulation ERCC1 and Rad51.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Emodina/farmacologia , Endonucleases/metabolismo , Extratos Vegetais/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Rad51 Recombinase/metabolismo , Análise de Variância , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Medicamentos de Ervas Chinesas/farmacologia , Endonucleases/efeitos dos fármacos , Endonucleases/genética , Expressão Gênica , Humanos , RNA Mensageiro/metabolismo , Rad51 Recombinase/efeitos dos fármacos , Rad51 Recombinase/genética , Vacúolos/ultraestrutura
20.
Methods Enzymol ; 504: 3-28, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22264527

RESUMO

The use of nano- and microbeam techniques to induce and identify subcellular localized energy deposition within a region of a living cell provides a means to investigate the effects of low radiation doses. Particularly within the nucleus where the propagation and processing of deoxyribonucleic acid (DNA) damage (and repair) in both targeted and nontargeted cells, the latter being able to study cell-cell (bystander) effects. We have pioneered a near infrared (NIR) femtosecond laser microbeam to mimic ionizing radiation through multiphoton absorption within a 3D femtoliter volume of a highly focused Gaussian laser beam. The novel optical microbeam mimics both complex ionizing and UV-radiation-type cell damage including double strand breaks (DSBs). Using the microbeam technology, we have been able to investigate the formation of DNA DSB and subsequent recruitment of repair proteins to the submicrometer size site of damage introduced in viable cells. The use of a phosphorylated H2AX (γ-H2AX a marker for DSBs, visualized by immunofluorescent staining) and real-time imaging of fluorescently labeling proteins, the dynamics of recruitment of repair proteins in viable mammalian cells can be observed. Here we show the recruitment of ATM, p53 binding protein 1 (53BP1), and RAD51, an integral protein of the homologous recombination process in the DNA repair pathway and Ku-80-GFP involved in the nonhomologous end joining (NHEJ) pathway as exemplar repair process to show differences in the repair kinetics of DNA DSBs. The laser NIR multiphoton microbeam technology shows persistent DSBs at later times post laser irradiation which are indicative of DSBs arising at replication presumably from UV photoproducts or clustered damage containing single strand breaks (SSBs) that are also observed. Effects of the cell cycle may also be investigated in real time. Postirradiation and fixed cells studies show that in G1 cells a fraction of multiphoton laser-induced DSBs is persistent for >6h in addition to those induced at replication demonstrating the broad range of timescales taken to repair DNA damage.


Assuntos
Células/citologia , Quebras de DNA de Cadeia Dupla , Histonas/análise , Lasers , Terapia com Luz de Baixa Intensidade/métodos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Animais , Antígenos Nucleares/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Efeito Espectador , Ciclo Celular/genética , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Células/efeitos da radiação , Reparo do DNA/efeitos da radiação , Replicação do DNA/genética , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Autoantígeno Ku , Mamíferos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Rad51 Recombinase/metabolismo , Radiação Ionizante , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA