Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 434
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-34202550

RESUMO

In this study, we determined the potential of polyethylene glycol-encapsulated iron oxide nanoparticles (IONPCO) for the intracellular delivery of the chemotherapeutic doxorubicin (IONPDOX) to enhance the cytotoxic effects of ionizing radiation. The biological effects of IONP and X-ray irradiation (50 kV and 6 MV) were determined in HeLa cells using the colony formation assay (CFA) and detection of γH2AX foci. Data are presented as mean ± SEM. IONP were efficiently internalized by HeLa cells. IONPCO radiomodulating effect was dependent on nanoparticle concentration and photon energy. IONPCO did not radiosensitize HeLa cells with 6 MV X-rays, yet moderately enhanced cellular radiosensitivity to 50 kV X-rays (DMFSF0.1 = 1.13 ± 0.05 (p = 0.01)). IONPDOX did enhance the cytotoxicity of 6 MV X-rays (DMFSF0.1 = 1.3 ± 0.1; p = 0.0005). IONP treatment significantly increased γH2AX foci induction without irradiation. Treatment of HeLa cells with IONPCO resulted in a radiosensitizing effect for low-energy X-rays, while exposure to IONPDOX induced radiosensitization compared to IONPCO in cells irradiated with 6 MV X-rays. The effect did not correlate with the induction of γH2AX foci. Given these results, IONP are promising candidates for the controlled delivery of DOX to enhance the cytotoxic effects of ionizing radiation.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos , Compostos Férricos , Nanopartículas Metálicas , Tolerância a Radiação/efeitos dos fármacos , Relação Dose-Resposta à Radiação , Portadores de Fármacos/química , Compostos Férricos/química , Células HeLa/efeitos dos fármacos , Células HeLa/patologia , Células HeLa/efeitos da radiação , Células HeLa/ultraestrutura , Humanos , Nanopartículas Metálicas/química , Radiação Ionizante
2.
Lasers Surg Med ; 51(3): 301-308, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30615224

RESUMO

BACKGROUND AND OBJECTIVE: Photodynamic therapy (PDT) has been widely used to treat malignant tumors. Our previous studies indicated that connexin (Cx) 32- and Cx26-composed gap junctional intercellular communication (GJIC) could improve the phototoxicity of PDT. However, the role of heterotypic Cx32/Cx26-formed GJIC in PDT phototoxicity is still unknown. Thus, the present study was aimed to investigate the effect of Cx32/Cx26-formed GJIC on PDT efficacy. METHODS: CCK8 assay was used to detect cell survival after PDT. Western blot assay was utilized to detect Cx32/Cx26 expression. "Parachute" dye-coupling assay was performed to measure the function of GJ channels. The intracellular Ca2+ concentrations were determined using flow cytometer. ELISA assay was performed to detect the intracellular levels of PGE2 and cAMP. RESULTS: The present study demonstrates there is a Cx32/Cx26-formed GJIC-dependent reduction of phototoxicity when cells were exposure to low concentration of Photofrin. Such a protective action is missing at low cell density due to the lack of GJ coupling. Under high-cell density condition, where there is opportunity for the cells to contact each other and form GJ, suppressing Cx32/Cx26-formed GJIC by either inhibiting the expression of Cx32/Cx26 or pretreating with GJ channel inhibitor augments PDT phototoxicity after cells were treated with at 2.5 µg/ml Photofrin. The above results suggest that at low Photofrin concentration, the presence of Cx32/Cx26-formed GJIC may decrease the phototoxicity of PDT, leading to the insensitivity of malignant cells to PDT treatment. The GJIC-mediated PDT insensitivity was associated with Ca2+ and prostaglandin E2 (PGE2 ) signaling pathways. CONCLUSION: The present study provides a cautionary note that for tumors expressing Cx32/Cx26, the presence of Cx32/Cx26-composed GJIC may cause the resistance of tumor cells to PDT. Oppositely, treatment strategies designed to downregulate the expression of Cx32/Cx26 or restrain the function of Cx32/Cx26-mediated GJIC may increase the sensitivity of malignant cell to PDT. Lasers Surg. Med. 51:301-308, 2019. © 2019 Wiley Periodicals, Inc.


Assuntos
Comunicação Celular/efeitos da radiação , Conexina 26/fisiologia , Conexinas/fisiologia , Junções Comunicantes/efeitos da radiação , Células HeLa/efeitos da radiação , Fotoquimioterapia/efeitos adversos , Técnicas de Cultura de Células , Sobrevivência Celular , Éter de Diematoporfirina/farmacologia , Células HeLa/patologia , Humanos , Fármacos Fotossensibilizantes/farmacologia , Proteína beta-1 de Junções Comunicantes
3.
PLoS One ; 12(11): e0186806, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29155820

RESUMO

Ultraviolet (UV) irradiation is a common form of DNA damage that can cause pyrimidine dimers between DNA, which can cause gene mutations, even double-strand breaks and threaten genome stability. If DNA repair systems default their roles at this stage, the organism can be damaged and result in disease, especially cancer. To better understand the cellular response to this form of damage, we applied highly sensitive mass spectrometry to perform comparative proteomics of phosphorylation in HeLa cells. A total of 4367 phosphorylation sites in 2100 proteins were identified, many of which had not been reported previously. Comprehensive bioinformatics analysis revealed that these proteins were involved in many important biological processes, including signaling, localization and cell cycle regulation. The nuclear pore complex, which is very important for RNA transport, was changed significantly at phosphorylation level, indicating its important role in response to UV-induced cellular stress. Protein-protein interaction network analysis and DNA repair pathways crosstalk were also examined in this study. Proteins involved in base excision repair, nucleotide repair and mismatch repair changed their phosphorylation pattern in response to UV treatment, indicating the complexity of cellular events and the coordination of these pathways. These systematic analyses provided new clues of protein phosphorylation in response to specific DNA damage, which is very important for further investigation. And give macroscopic view on an overall phosphorylation situation under UV radiation.


Assuntos
Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Dímeros de Pirimidina/efeitos da radiação , Células HeLa/efeitos da radiação , Humanos , Espectrometria de Massas , Fosforilação/efeitos da radiação , Mapas de Interação de Proteínas/efeitos da radiação , Raios Ultravioleta
4.
Sci Rep ; 7(1): 10919, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28883430

RESUMO

We report on a highly efficient magneto-actuated cancer cell apoptosis method using a biaxial pulsed magnetic field configuration, which maximizes the induced magnetic torque. The light transmissivity dynamics show that the biaxial magnetic field configuration can actuate the magnetic nanoparticles with higher responsiveness over a wide range of frequencies as compared to uniaxial field configurations. Its efficacy was demonstrated in in vitro cell destruction experiments with a greater reduction in cell viability. Magnetic nanoparticles with high aspect ratios were also found to form a triple vortex magnetization at remanence which increases its low field susceptibility. This translates to a larger magneto-mechanical actuated force at low fields and 12% higher efficacy in cell death as compared to low aspect ratio nanoparticles.


Assuntos
Apoptose/efeitos da radiação , Células HeLa/fisiologia , Células HeLa/efeitos da radiação , Campos Magnéticos , Nanopartículas Metálicas/efeitos da radiação , Humanos
5.
Zhonghua Zhong Liu Za Zhi ; 38(10): 725-730, 2016 Oct 23.
Artigo em Chinês | MEDLINE | ID: mdl-27784453

RESUMO

Objective: To investigate the killing effect of low-temperature plasma (LTP) on HepG2, A549 and HeLa cell lines and explore its possible mechanism. Methods: The inhibitory effect of LTP on the proliferation of HepG2, A549 and HeLa cells was determined by MTT assay. Transmission electron microscopy was used to observe the ultrastructural changes of HepG2, A549 and HeLa cells treated with LTP. Cell apoptosis was detected by Muse cytometry. Western blot was used to detect the expression of apoptosis-related proteins. Results: The survival rates of LTP-irradiated HepG2 cells (irradiated for 107 s), HeLa cells (irradiated for 121 s) and A549 cells (irradiated for 127 s) were 50%. LTP destroyed the ultrastructure of HepG2, A549 and HeLa cells to different degrees, showing nuclear fragmentation and organelle damages. The apoptosis rates of the three cell lines were increased at 24 h after exposure to LTP for 1/6 IC50 irradiation time. Furthermore, LTP irradiation also suppressed the protein expression of Bcl-2 and XRCC1 and increased that of Bax. Conclusions: LTP has an obvious killing effect on HepG2, A549 and HeLa cancer cell lines. This effect may be related to the induction of cell apoptosis and inhibition of DNA repair.


Assuntos
Células A549/fisiologia , Apoptose , Proliferação de Células , Crioterapia/métodos , Células HeLa/fisiologia , Células Hep G2/fisiologia , Células A549/efeitos da radiação , Células A549/ultraestrutura , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células HeLa/efeitos da radiação , Células HeLa/ultraestrutura , Células Hep G2/efeitos da radiação , Células Hep G2/ultraestrutura , Humanos
6.
Cold Spring Harb Protoc ; 2016(7)2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27371593

RESUMO

Ultraviolet (UV) radiation is a convenient stimulus for triggering cell death that is available in most laboratories. We use a Stratalinker UV cross-linker because it is a safe, cheap, reliable, consistent, and easily controlled source of UV irradiation. This protocol describes using a Stratalinker to trigger UV-induced death of HeLa cells.


Assuntos
Morte Celular , Células HeLa/efeitos da radiação , Raios Ultravioleta , Humanos
7.
DNA Cell Biol ; 35(3): 140-5, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26717101

RESUMO

The tumor suppressor, cylindromatosis (CYLD), is a negative regulator of NF-κB signaling by removing lysine 63-linked ubiquitin chains from multiple NF-κB signaling components, including TRAF2, TRAF6, and NEMO. How CYLD itself is regulated, however, remains yet to be characterized. In this study, we present the first evidence that UV irradiation is able to induce CYLD translocation from the cytoplasm to microtubules and that the cytoskeleton-associated CYLD is subject to posttranslational modification and degradation in a proteasome-independent manner. By immunostaining, we found that CYLD displayed microtubule-like filament localization under ultraviolet (UV) irradiation. Further studies revealed that the cytoskeleton-associated CYLD underwent posttranslational modification, which in turn contributed to CYLD degradation in an unknown manner, distinct from proteasome-mediated degradation under normal conditions. Collectively, our data suggest that UV-induced CYLD degradation might serve as an underlying mechanism for UV-induced NF-κB pathway activation.


Assuntos
Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Raios Ultravioleta/efeitos adversos , Citoplasma/metabolismo , Citoplasma/efeitos da radiação , Citoesqueleto/metabolismo , Enzima Desubiquitinante CYLD , Células HeLa/efeitos da radiação , Humanos , Células MCF-7/efeitos da radiação , Microtúbulos/metabolismo , Microtúbulos/efeitos da radiação , Processamento de Proteína Pós-Traducional/efeitos da radiação , Transporte Proteico/efeitos da radiação
8.
Acta Biochim Pol ; 62(2): 173-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26015993

RESUMO

Histone modifications are involved in the DNA damage response (DDR). Here, by utilizing an ELISA immunoassay we assessed the methylation at H3K9 (H3K9me2 and H3K9me3) in two cell lines with differential sensitivity to radiation-induced apoptosis, HeLa (sensitive) and MCF-7 (resistant). We found that DNA damage induction by γ-irradiation leads to considerable accumulation (up to 5-fold) of H3K9me2 and H3K9me3, but not of H4K20me3 (control modification) in MCF-7 cells (p<0.05). Interestingly, a lower dose (2 Gy) was more effective than 5 Gy. In HeLa cells a smaller effect (approx. 1.5-1.8-fold) was evident only at 5 Gy. In conclusion, our findings reveal that DNA damage leads to specific accumulation of H3K9me2 and H3K9me3 in a cell-type specific manner.


Assuntos
Heterocromatina/metabolismo , Histonas/metabolismo , Radiação Ionizante , Dano ao DNA/fisiologia , Dano ao DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Células HeLa/metabolismo , Células HeLa/efeitos da radiação , Heterocromatina/efeitos da radiação , Humanos , Lisina/metabolismo , Células MCF-7/metabolismo , Células MCF-7/efeitos da radiação , Metilação , Tolerância a Radiação , Proteína Supressora de Tumor p53/metabolismo
9.
Genes Cells ; 20(3): 191-202, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25482373

RESUMO

In eukaryotes, holo-Mediator consists of four modules: head, middle, tail, and CDK/Cyclin. The head module performs an essential function involved in regulation of RNA polymerase II (Pol II). We studied the human head module subunit MED17 (hMED17). Recent structural studies showed that yeast MED17 may function as a hinge connecting the neck and movable jaw regions of the head module to the fixed jaw region. Luciferase assays in hMED17-knockdown cells showed that hMED17 supports transcriptional activation, and pulldown assays showed that hMED17 interacted with Pol II and the general transcription factors TFIIB, TBP, TFIIE, and TFIIH. In addition, hMED17 bound to a DNA helicase subunit of TFIIH, XPB, which is essential for both transcription and nucleotide excision repair (NER). Because hMED17 associates with p53 upon UV-C irradiation, we treated human MCF-7 cells with either UV-C or the MDM2 inhibitor Nutlin-3. Both treatments resulted in accumulation of p53 in the nucleus, but hMED17 remained concentrated in the nucleus in response to UV-C. hMED17 colocalized with the NER factors XPB and XPG following UV-C irradiation, and XPG and XPB bound to hMED17 in vitro. These findings suggest that hMED17 may play essential roles in switching between transcription and NER.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Complexo Mediador/metabolismo , Fatores de Transcrição/metabolismo , Inibidores Enzimáticos/farmacologia , Células HeLa/efeitos da radiação , Humanos , Imidazóis/farmacologia , Células MCF-7/efeitos dos fármacos , Células MCF-7/efeitos da radiação , Complexo Mediador/genética , Piperazinas/farmacologia , Ligação Proteica , Transporte Proteico/efeitos da radiação , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Polimerase II/metabolismo , Ativação Transcricional , Raios Ultravioleta
10.
Radiat Res ; 181(5): 540-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24785588

RESUMO

Crosstalk between cancer cells and the surrounding cancer associated fibroblasts (CAFs) plays an illusive role in cancer radiotherapy. This study investigated the effect of cancer cell-cancer associated fibroblasts crosstalk on the proliferation and survival of irradiated cervical cancer cells. A pretreatment with conditioned medium from a mixed culture of CAF and HeLa cells (mixCAF) had a stronger effect on enhancing the proliferation and survival of irradiated HeLa cells compared to pretreatment with CAF conditioned medium alone. In addition, pretreatment with a mixed culture of CAF and HeLa cells conditioned medium reduced the levels of two major radiation-induced genes, GADD45 and BTG2, and phosphorylation of p38. Profiling of the growth and survival factors in the conditioned medium revealed PDGF and VEGF, and IGF2, EGF, FGF-4, IGFBPs and GM-CSF to be specifically secreted from HeLa cells and CAFs, respectively. This study demonstrated radiation protective effects of CAF-cancer cell crosstalk, and identified multiple growth factors and radiation response genes that might be involved in these effects.


Assuntos
Carcinoma de Células Escamosas/patologia , Comunicação Celular , Células Epiteliais/patologia , Fibroblastos/patologia , Células Estromais/patologia , Microambiente Tumoral , Neoplasias do Colo do Útero/patologia , Animais , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos da radiação , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Feminino , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células HeLa/metabolismo , Células HeLa/efeitos da radiação , Células HeLa/transplante , Humanos , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Fosforilação/efeitos da radiação , Processamento de Proteína Pós-Traducional/efeitos da radiação , Tolerância a Radiação , Reação em Cadeia da Polimerase em Tempo Real , Células Estromais/metabolismo , Ensaio Tumoral de Célula-Tronco , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Bioorg Med Chem ; 22(3): 1003-7, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24417957

RESUMO

We developed a novel method for regulation of RISC function by photoreactive oligonucleotides (Ps-Oligo) containing 2'-O-psoralenylmethoxyethyl adenosine (Aps). We observed that inhibitory effects of Ps-Oligos on RISC function were enhanced by UV-irradiation compared with 2'-O-methyl-oligonucleotide without Aps. These results suggest Ps-Oligo inhibited RISC function by cross-linking effect, and we propose that the concept described in this report may be promising and applicable one to regulate the small RNA-mediated post-transcriptional regulation.


Assuntos
Ficusina/química , Oligonucleotídeos/química , Oligonucleotídeos/farmacocinética , Complexo de Inativação Induzido por RNA/genética , Adenosina , Reagentes de Ligações Cruzadas/química , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Oligonucleotídeos Antissenso/farmacologia , Fotoquímica/métodos , Complexo de Inativação Induzido por RNA/química , Termodinâmica , Thermus thermophilus/genética , Raios Ultravioleta
12.
Radiats Biol Radioecol ; 54(3): 256-64, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25764829

RESUMO

Radioresistance of cancer stem cells (CSCs) is regarded as one of the possible causes of cancer recurrence after radiotherapy. Since the regularities and mechanisms of radiation effects on this population of cells have not been sufficiently studied, the aim of this work is to elucidate the changes in the CSC number after γ-irradiation in stable cultures of tumor cells in vitro and tumor tissue in vivo (in the course of radiation therapy of patients with cancers of the upper respiratory tract). CSCs were identified in the cell lines B16, MCF-7, HeLa by the ability to exclude the fluorescent dye Hoechst 33342 (SP method) 48-72 h after irradiation at the doses of 1-20 Gy and in biopsy material by immunophenotype CD44+CD24(-/low) before and 24 h after irradiation at the total dose of 10 Gy. The essential differences in the response of CSCs and other cancer cells were found after exposure to low-LET radiation. The absolute number of CSCs increased after a single exposure at the doses ranging from 1 to 5-10 Gy in different cell cultures, but a further dose increase maintained the current number of CSCs or decreased it. At the same time, the number of non CSCs significantly decreased with increasing doses of radiation exposure, as expected. Fractionated irradiation in vivo at a total dose of 10 Gy increased the relative amount of CSCs in most patients. The registered changes are an integral indicator of cell death, cell division delay immediately after irradiation, proliferation at a later time, possible dedifferentiation of non CSCs, etc. The exact contribution of each of them to the radiation-induced increase of the CSCs number is of considerable interest and requires further research.


Assuntos
Raios gama , Neoplasias Laríngeas/radioterapia , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação/genética , Adulto , Idoso , Animais , Feminino , Células HeLa/efeitos da radiação , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Imunofenotipagem , Neoplasias Laríngeas/patologia , Células MCF-7/efeitos da radiação , Masculino , Camundongos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/ultraestrutura
13.
Arch Gynecol Obstet ; 289(5): 1101-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24292149

RESUMO

OBJECTIVE: To compare the efficacies and side effects of Iodine-125 (125I) seeds implantation with afterloading radiotherapy on tumor xenografts derived from Hela cells. METHODS: Mice bearing Hela cells were randomly divided into three groups: two therapeutic groups receiving four 125I seeds implantation and afterloading therapy, respectively, and the one control group without any intervention. Comparisons were evaluated in aspects of curative efficacies (tumor volume, tumor inhibition rate and metastasis) and side effects (body weight, ovarian endocrine functions, skin lesion surrounding the tumor). RESULTS: The xenografts tumor volume of therapeutic groups were significantly smaller than that of controls(p < 0.05),both of the 125I seeds implantation and afterloading therapy showed excellent tumor inhibition rate. Furthermore, 125I seeds implantation had milder side effects on skin, weight loss, ovarian endocrine functions. CONCLUSIONS: (125)I seed implantation may be an alternative minimally invasive therapy for cervical cancer.


Assuntos
Braquiterapia/métodos , Radioisótopos do Iodo/uso terapêutico , Neoplasias do Colo do Útero/radioterapia , Animais , Feminino , Células HeLa/efeitos da radiação , Xenoenxertos/efeitos da radiação , Humanos , Iodo , Camundongos , Camundongos Endogâmicos BALB C , Dosagem Radioterapêutica , Distribuição Aleatória , Resultado do Tratamento
14.
Mol Cell Biol ; 33(11): 2212-27, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23530059

RESUMO

FANCJ mutations are linked to Fanconi anemia (FA) and increase breast cancer risk. FANCJ encodes a DNA helicase implicated in homologous recombination (HR) repair of double-strand breaks (DSBs) and interstrand cross-links (ICLs), but its mechanism of action is not well understood. Here we show with live-cell imaging that FANCJ recruitment to laser-induced DSBs but not psoralen-induced ICLs is dependent on nuclease-active MRE11. FANCJ interacts directly with MRE11 and inhibits its exonuclease activity in a specific manner, suggesting that FANCJ regulates the MRE11 nuclease to facilitate DSB processing and appropriate end resection. Cells deficient in FANCJ and MRE11 show increased ionizing radiation (IR) resistance, reduced numbers of γH2AX and RAD51 foci, and elevated numbers of DNA-dependent protein kinase catalytic subunit foci, suggesting that HR is compromised and the nonhomologous end-joining (NHEJ) pathway is elicited to help cells cope with IR-induced strand breaks. Interplay between FANCJ and MRE11 ensures a normal response to IR-induced DSBs, whereas FANCJ involvement in ICL repair is regulated by MLH1 and the FA pathway. Our findings are discussed in light of the current model for HR repair.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Dano ao DNA , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Hidrolases Anidrido Ácido , Fatores de Transcrição de Zíper de Leucina Básica/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Instabilidade Cromossômica , Quebras de DNA de Cadeia Dupla , Reparo do DNA/efeitos da radiação , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Endodesoxirribonucleases , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Ficusina/farmacologia , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Proteína Homóloga a MRE11 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Radiação Ionizante , Reparo de DNA por Recombinação
15.
Mol Cell ; 45(6): 801-13, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22361354

RESUMO

The deubiquitylation enzyme USP7/HAUSP plays a major role in regulating genome stability and cancer prevention by controlling the key proteins involved in the DNA damage response. Despite this important role in controlling other proteins, USP7 itself has not been recognized as a target for regulation. Here, we report that USP7 regulation plays a central role in DNA damage signal transmission. We find that stabilization of Mdm2, and correspondingly p53 downregulation in unstressed cells, is accomplished by a specific isoform of USP7 (USP7S), which is phosphorylated at serine 18 by the protein kinase CK2. Phosphorylation stabilizes USP7S and thus contributes to Mdm2 stabilization and downregulation of p53. After ionizing radiation, dephosphorylation of USP7S by the ATM-dependent protein phosphatase PPM1G leads to USP7S downregulation, followed by Mdm2 downregulation and accumulation of p53. Our findings provide a quantitative transmission mechanism of the DNA damage signal to coordinate a p53-dependent DNA damage response.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina Tiolesterase/metabolismo , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Células HeLa/efeitos da radiação , Humanos , Dados de Sequência Molecular , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Fosfatase 2C , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Radiação Ionizante , Serina/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética , Peptidase 7 Específica de Ubiquitina
16.
J Radiat Res ; 53(1): 58-71, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22302046

RESUMO

Reactive oxygen species (ROS) act as a mediator of ionizing radiation-induced cellular damage. Previous studies have indicated that MnSOD (SOD2) plays a critical role in protection against ionizing radiation in mammalian cells. In this study, we constructed two types of stable HeLa cell lines overexpressing SOD2, HeLa S3/SOD2 and T-REx HeLa/SOD2, to elucidate the mechanisms underlying the protection against radiation by SOD2. SOD2 overexpression in mitochondria enhanced the survival of HeLa S3 and T-REx HeLa cells following γ-irradiation. The levels of γH2AX significantly decreased in HeLa S3/SOD2 and T-REx HeLa/SOD2 cells compared with those in the control cells. MitoSox(TM) Red assays showed that both lines of SOD2-expressing cells showed suppression of the superoxide generation in mitochondria. Furthermore, flow cytometry with a fluorescent probe (2',7'-dichlorofluorescein) revealed that the cellular levels of ROS increased in HeLa S3 cells during post-irradiation incubation, but the increase was markedly attenuated in HeLa S3/SOD2 cells. DNA microarray analysis revealed that, of 47,000 probe sets analyzed, 117 and 166 probes showed more than 2-fold changes after 5.5 Gy of γ-irradiation in control and HeLa S3/SOD2 cells, respectively. Pathway analysis revealed different expression profiles in irradiated control cells and irradiated SOD2-overexpressing cells. These results indicate that SOD2 protects HeLa cells against cellular effects of γ-rays through suppressing oxidative stress in irradiated cells caused by ROS generated in the mitochondria and through regulating the expression of genes which play a critical role in protection against ionizing radiation.


Assuntos
Regulação da Expressão Gênica/efeitos da radiação , Proteínas de Neoplasias/fisiologia , Tolerância a Radiação/fisiologia , Superóxido Dismutase/fisiologia , Quebras de DNA de Cadeia Dupla , Indução Enzimática , Raios gama/efeitos adversos , Redes Reguladoras de Genes/efeitos da radiação , Células HeLa/metabolismo , Células HeLa/efeitos da radiação , Humanos , Mitocôndrias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/fisiologia , Estresse Oxidativo/efeitos da radiação , Tolerância a Radiação/genética , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Superóxidos/metabolismo
17.
J Radiat Res ; 52(4): 481-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21785236

RESUMO

To understand how human tumor cells respond to the combined treatment with nocodazole and high LET radiation, alterations in cell cycle, mitotic disturbances and cell death were investigated in the present study. Human cervix carcinoma HeLa cells were exposed to nocodazole for 18 h immediately followed by high LET iron ion irradiation and displayed a sequence of events leading to DNA damages, mitotic aberrations, interphase restitution and endocycle as well as cell death. A prolonged mitotic arrest more than 10 h was observed following nocodazole exposure, no matter the irradiation was present or not. The occurrence of mitotic slippage following the mitotic arrest was only drug-dependent and the irradiation did not accelerate it. The amount of polyploidy cells was increased following mitotic slippage. No detectable G(2) or G(1) arrest was observed in cells upon the combined treatment and the cells reentered the cell cycle still harboring unrepaired cellular damages. This premature entry caused an increase of multipolar mitotic spindles and amplification of centrosomes, which gave rise to lagging chromosomal material, failure of cytokinesis and polyploidization. These mitotic disturbances and their outcomes confirmed the incidence of mitotic catastrophe and delayed apoptotic features displayed by TUNEL method after the combined treatment. These results suggest that the addition of high-LET iron ion irradiation to nocodazole enhanced mitotic catastrophe and delayed apoptosis in HeLa cells. These might be important cell death mechanisms involved in tumor cells in response to the treatment of antimitotic drug combined with high LET radiation.


Assuntos
Apoptose/efeitos da radiação , Células HeLa/efeitos da radiação , Transferência Linear de Energia , Mitose/efeitos da radiação , Nocodazol/farmacologia , Antineoplásicos/farmacologia , Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Dano ao DNA , Humanos , Marcação In Situ das Extremidades Cortadas , Poliploidia , Fuso Acromático , Tubulina (Proteína)/química
18.
Radiother Oncol ; 101(1): 66-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21641068

RESUMO

BACKGROUND: Tumor radioresistance often leads to treatment failure during radiotherapy. New strategies like developing radiosensitizer are clinically important. Intervention with DNA double-strand break repair is an effective way to modulate tumor cell radiosensitivity. This study focused on the mutant Artemis fragment-enhanced radiosensitivity of human cervical cancer cells. MATERIAL AND METHODS: We constructed two pEGFP-C1-based eukaryotic expression vectors encoding full-length and the mutant Artemis fragment (D37N-413aa), respectively. HeLa cells were stably transfected with these plasmids or vector. Cell survival was measured by the clonogenic assay. The γH2AX foci assay was used to monitor DNA repair after irradiation. Co-immunoprecipitation and Western blot analysis were performed to study protein interaction and phosphorylation of Artemis. RESULTS: Expression of the mutant Artemis fragment (D37N-413aa) delayed DNA DSB rejoining after irradiation, thereby enhanced radiosensitivity of HeLa cell. Further experiments indicate that this mutant Artemis fragment bind to DNA-PKcs and ATM, inhibited phosphorylation of endogenous Artemis, the key molecule for DNA repair and cell radiosensitivity. CONCLUSIONS: The dominant negative mutant Artemis fragment (D37N-413aa) enhanced tumor cell radiosensitivity through blocking activity of endogenous Artemis and DNA repair. It is the first time to modulate tumor cell radiosensitivity via targeting Artemis. This novel mechanism of radiosensitivity strongly suggests the potential role of Artemis in cancer therapy.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Proteínas Mutantes Quiméricas/farmacologia , Proteínas Nucleares/genética , Tolerância a Radiação/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Endonucleases , Feminino , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Proteínas Mutantes Quiméricas/genética , Fosforilação , Tolerância a Radiação/genética , Sensibilidade e Especificidade , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/farmacologia
19.
Appl Biochem Biotechnol ; 163(8): 1002-11, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20953732

RESUMO

Exposure of mammalian cells to oxidative stress alters lysosomal enzymes. Through cytochemical analysis of lysosomes with LysoTracker, we demonstrated that the number and fluorescent intensity of lysosome-like organelles in HeLa cells increased with exposure to hydrogen peroxide (H2O2), 6-hydroxydopamine (6-OHDA), and UVB irradiation. The lysosomes isolated from HeLa cells exposed to three oxidative stressors showed the enhanced antimicrobial activity against Escherichia coli. Further, when lysosomes that were isolated from HeLa cells exposed by oxidative stress were treated to normal HeLa cells, the viability of the HeLa cells was drastically reduced, suggesting increased in vitro lysosomal function (i.e., antimicrobial activity, apoptotic cell death). In addition, we also found that cathepsin B and D were implicated in increased in vitro lysosomal function when isolated from HeLa cells exposed by oxidative stress. Decrease in cathepsin B activity and increase in cathepsin D activity were observed in lysosomes isolated from HeLa cells after treatment with H2O2, 6-ODHA, or UVB, but cathepsin B and D were not the sole factors to induce cell death by in vitro lysosomal function. Therefore, these studies suggest a new approach to use lysosomes as antimicrobial agents and as new materials for treating cancer cell lines.


Assuntos
Células HeLa/metabolismo , Lisossomos/metabolismo , Estresse Oxidativo , Apoptose , Catepsina B/metabolismo , Catepsina D/metabolismo , Células HeLa/citologia , Células HeLa/efeitos dos fármacos , Células HeLa/efeitos da radiação , Humanos , Peróxido de Hidrogênio/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/efeitos da radiação , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Raios Ultravioleta
20.
J Cell Physiol ; 226(2): 369-74, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20665702

RESUMO

Low-dose (≤0.1 Gy) radiation-induced adaptive responses could protect cells from high-challenge dose radiation-induced killing. The protective role is believed to promote the repair of DNA double-strand breaks (DSBs) that are a severe threat to cell survival. However, it remains unclear which repair pathway, homologous recombination repair (HRR) or non-homologous end-joining (NHEJ), is promoted by low-dose radiation. To address this question, we examined the effects of low-dose (0.1 Gy) on high-challenge dose (2-4 Gy) induced killing in NHEJ- or HRR-deficient cell lines. We showed that 0.1 Gy reduced the high-dose radiation-induced killing for wild-type or HRR-deficient cells, but enhanced the killing for NHEJ-deficient cells. Interestingly, low-dose radiation also enhanced the killing for wild-type cells exposed to high-challenge dose radiation with high-linear energy transfer (LET). Because it is known that high-LET radiation induces an inefficient NHEJ, these results support that the low-dose radiation-stimulated protective role in reducing high-challenge dose (low-LET)-induced cell killing might depend on NHEJ. In addition, we showed that low-dose radiation activated the DNA-PK catalytic subunit (DNA-PKcs) and the inhibitor of DNA-PKcs destroyed the low-dose radiation-induced protective role. These results suggest that low-dose radiation might promote NHEJ through the stimulation of DNA-PKcs activity and; therefore, increase the resistance of cells to high-challenge dose radiation-induced killing.


Assuntos
Antígenos Nucleares/metabolismo , Sobrevivência Celular/efeitos da radiação , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Recombinação Genética , Animais , Antígenos Nucleares/genética , Células CHO/efeitos da radiação , Cricetinae , Cricetulus , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Células HeLa/efeitos da radiação , Humanos , Autoantígeno Ku , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA