Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 418
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Bull Exp Biol Med ; 174(4): 489-496, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36899199

RESUMO

We explored the mechanism by which miR-139 modulates radioresistance of esophageal cancer (EC). The radioresistant cell line KYSE150R was obtained from the parental KYSE150 cell line by fractionated irradiation (15×2 Gy; total dose of 30 Gy). The cell cycle was assessed by flow cytometry. A gene profiling study was conducted to detect the expression of genes related to the radioresistance of EC. In the KYSE150R line, flow cytometry revealed increased number of G1-phase cells and decreased number of G2-phase cells; the expression of miR-139 increased. Knockdown of miR-139 decreased radioresistance and changed the distribution of cell cycle phases in KYSE150R cells. Western blotting showed that miR-139 knockdown increased the expression levels of cyclin D1, p-AKT, and PDK1. However, PDK1 inhibitor GSK2334470 reversed this effect for p-AKT and cyclin D1 expression. A luciferase reporter assay indicated that miR-139 directly bound to the PDK1 mRNA 3'-UTR. Analysis of the clinical data from 110 patients with EC showed an association of miR-139 expression with the TNM stage and the effect of therapy. MiR-139 expression significantly correlated with EC and progression-free survival. In conclusion, miR-139 enhances the radiosensitivity of EC by regulating the cell cycle through the PDK1/Akt/Cyclin D1 signaling pathway.


Assuntos
Neoplasias Esofágicas , MicroRNAs , Tolerância a Radiação , Humanos , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Linhagem Celular Tumoral/efeitos da radiação , Proliferação de Células/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/radioterapia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Radiação/genética , Transdução de Sinais/genética
2.
Gynecol Obstet Invest ; 86(1-2): 71-80, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33260174

RESUMO

OBJECTIVE: The objective of this study is to explore the role of miR-210 in the growth of ovarian cancer cells and the correlation with radiotherapy and to elucidate underlying molecular mechanisms. METHODS: Human ovarian cancer cell lines OVCAR3 and SKOV3 were cultured in vitro, and miR-210 over-expression and low-expression ovarian cancer cell models were established by cell transfection. MTT assay was used to detect the proliferation activity. Transwell was used to detect the migration and invasion abilities. Western blot measured the expression of proteins related to cell proliferation, migration, and invasion. The cells were treated with different doses of ionizing radiation, and then the cell proliferation activity was detected by MTT. The expression of apoptosis-related proteins was detected by Western blot. The Caspase-Glo® Kit was used to detect the activity of cellular caspase 3/7 enzymes. RESULTS: The proliferation, migration, and invasion abilities of miR-210 over-expression ovarian cancer cells were increased (p < 0.05), the expressions of PTEN and E-cadherin were decreased, and the expression of p-Protein kinase B (AKT), N-cadherin, Snail, and Vimentin were elevated. After ionizing radiation, the sensitivity of miR-210 over-expression cells to radiotherapy was decreased, the expression of apoptosis-related protein Bax was decreased, the expression of Bcl-2 was increased, and the activity of cellular caspase 3/7 enzyme was reduced (p < 0.05). CONCLUSION: miR-210 can promote the proliferation, migration, and invasion of ovarian cancer cells by activating the AKT signaling pathway and regulating the expression of Epithelial-mesenchymal transition-related proteins. miR-210 can reduce the sensitivity of ovarian cancer cells to radiotherapy by inhibiting apoptosis, which might serve as a potential target for the treatment of ovarian tumors.


Assuntos
Linhagem Celular Tumoral/efeitos da radiação , MicroRNAs/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/radioterapia , Apoptose , Caderinas/metabolismo , Movimento Celular , Proliferação de Células , Feminino , Humanos , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Transdução de Sinais , Vimentina/metabolismo , Proteína X Associada a bcl-2
3.
J Biomed Sci ; 27(1): 90, 2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32854690

RESUMO

BACKGROUND: The probability of local tumor control after radiotherapy (RT) remains still miserably poor in pediatric rhabdomyosarcoma (RMS). Thus, understanding the molecular mechanisms responsible of tumor relapse is essential to identify personalized RT-based strategies. Contrary to what has been done so far, a correct characterization of cellular radioresistance should be performed comparing radioresistant and radiosensitive cells with the same isogenic background. METHODS: Clinically relevant radioresistant (RR) embryonal (RD) and alveolar (RH30) RMS cell lines have been developed by irradiating them with clinical-like hypo-fractionated schedule. RMS-RR cells were compared to parental isogenic counterpart (RMS-PR) and studied following the radiobiological concept of the "6Rs", which stand for repair, redistribution, repopulation, reoxygenation, intrinsic radioresistance and radio-immuno-biology. RESULTS: RMS-RR cell lines, characterized by a more aggressive and in vitro pro-metastatic phenotype, showed a higher ability to i) detoxify from reactive oxygen species; ii) repair DNA damage by differently activating non-homologous end joining and homologous recombination pathways; iii) counteract RT-induced G2/M cell cycle arrest by re-starting growth and repopulating after irradiation; iv) express cancer stem-like profile. Bioinformatic analyses, performed to assess the role of 41 cytokines after RT exposure and their network interactions, suggested TGF-ß, MIF, CCL2, CXCL5, CXCL8 and CXCL12 as master regulators of cancer immune escape in RMS tumors. CONCLUSIONS: These results suggest that RMS could sustain intrinsic and acquire radioresistance by different mechanisms and indicate potential targets for future combined radiosensitizing strategies.


Assuntos
Linhagem Celular Tumoral/efeitos da radiação , Tolerância a Radiação , Rabdomiossarcoma Alveolar/radioterapia , Rabdomiossarcoma Embrionário/radioterapia , Humanos
4.
Breast J ; 26(1): 47-54, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31944484

RESUMO

While there is now Level I data with long-term follow-up supporting the routine use of hypofractionated (HF) whole-breast radiation therapy (WBRT) after breast-conserving surgery, its adoption has been slow and variable. This article will review the literature supporting the efficacy and safety of hypofractionated radiation for breast cancer, discuss the radiobiological rationale specific to breast tumors, and make an argument for justifying the routine adoption of shorter, HF-WBRT courses when delivering breast radiation. Data using HF with regional nodal irradiation and in the post-mastectomy setting will also be reviewed. The aim is to provide an in-depth understanding of the use of hypofractionated radiation therapy for breast cancer, its applicability, and topics warranting future research.


Assuntos
Neoplasias da Mama/radioterapia , Hipofracionamento da Dose de Radiação , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/efeitos da radiação , Feminino , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
5.
BMC Genomics ; 20(1): 611, 2019 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-31345182

RESUMO

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) cells are heterogeneous, easily develop radioresistance, and recur. Single-cell RNA-seq (scRNA-seq) is a next-generation sequencing method that can delineate diverse gene expression profiles of individual cells and mining their heterogeneous behaviors in response to irradiation. Our aim was using scRNA-seq to describe the difference between parental cells and cells that acquired radioresistance, and to investigate the dynamic changes of the transcriptome of cells in response to FIR. RESULTS: We sequenced ESCC cell lines KYSE180 with and without fractionated irradiation (FIR). A total of 218 scRNA-seq libraries were obtained from 88 cells exposed to 12 Gy (KYSE-180-12 Gy), 89 exposed to 30 Gy (KYSE-180-30 Gy), and 41 parental KYSE-180 cells not exposed to FIR. Dynamic gene expression patterns were determined by comprehensive consideration of genes and pathways. Biological experiments showed that KYSE-180 cells became radioresistant after FIR. PCA analysis of scRNA-seq data showed KYSE-180, KYSE-180-12 Gy and KYSE-180-30 Gy cells were discrete away from each other. Two sub-populations found in KYSE-180-12 Gy and only one remained in KYSE-180-30 Gy. This sub-population genes exposure to FIR through 12 Gy to 30 Gy were relevant to the PI3K-AKT pathway, pathways evading apoptosis, tumor cell migration, metastasis, or invasion pathways, and cell differentiation and proliferation pathways. We validated DEGs, such as CFLAR, LAMA5, ITGA6, ITGB4, and SDC4 genes, in these five pathways as radioresistant genes in bulk cell RNA-seq data from ESCC tissue of a ESCC patient treated with radiotherapy and from KYSE-150 cell lines. CONCLUSIONS: Our results delineated the divergent gene expression patterns of individual ESCC cells exposure to FIR, and displayed genes and pathways related to development of radioresistance.


Assuntos
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Tolerância a Radiação , Transcriptoma , Linhagem Celular Tumoral/efeitos da radiação , Humanos , Redes e Vias Metabólicas , RNA-Seq
6.
Breast Cancer Res ; 20(1): 126, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348199

RESUMO

BACKGROUND: Although metastasis is ultimately responsible for about 90% of breast cancer mortality, the vast majority of breast-cancer-related deaths are due to progressive recurrences from non-metastatic disease. Current adjuvant therapies are unable to prevent progressive recurrences for a significant fraction of patients with breast cancer. Autologous tumor cell vaccines (ATCVs) are a safe and potentially useful strategy to prevent breast cancer recurrence, in a personalized and patient-specific manner, following standard-of-care tumor resection. Given the high intra-patient and inter-patient heterogeneity in breast cancer, it is important to understand which factors influence the immunogenicity of breast tumor cells in order to maximize ATCV effectiveness. METHODS: The relative immunogenicity of two murine breast carcinomas, 4T1 and EMT6, were compared in a prophylactic vaccination-tumor challenge model. Differences in cell surface expression of antigen-presentation-related and costimulatory molecules were compared along with immunosuppressive cytokine production. CRISPR/Cas9 technology was used to modulate tumor-derived cytokine secretion. The impacts of cytokine deletion on splenomegaly, myeloid-derived suppressor cell (MDSC) accumulation and ATCV immunogenicity were assessed. RESULTS: Mice vaccinated with an EMT6 vaccine exhibited significantly greater protective immunity than mice vaccinated with a 4T1 vaccine. Hybrid vaccination studies revealed that the 4T1 vaccination induced both local and systemic immune impairments. Although there were significant differences between EMT6 and 4T1 in the expression of costimulatory molecules, major disparities in the secretion of immunosuppressive cytokines likely accounts for differences in immunogenicity between the cell lines. Ablation of one cytokine in particular, granulocyte-colony stimulating factor (G-CSF), reversed MDSC accumulation and splenomegaly in the 4T1 model. Furthermore, G-CSF inhibition enhanced the immunogenicity of a 4T1-based vaccine to the extent that all vaccinated mice developed complete protective immunity. CONCLUSIONS: Breast cancer cells that express high levels of G-CSF have the potential to diminish or abrogate the efficacy of breast cancer ATCVs. Fortunately, this study demonstrates that genetic ablation of immunosuppressive cytokines, such as G-CSF, can enhance the immunogenicity of breast cancer cell-based vaccines. Strategies that combine inhibition of immunosuppressive factors with immune stimulatory co-formulations already under development may help ATCVs reach their full potential.


Assuntos
Neoplasias da Mama/terapia , Vacinas Anticâncer/imunologia , Fator Estimulador de Colônias de Granulócitos/imunologia , Imunogenicidade da Vacina , Recidiva Local de Neoplasia/prevenção & controle , Animais , Mama/patologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Sistemas CRISPR-Cas , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral/imunologia , Linhagem Celular Tumoral/efeitos da radiação , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Feminino , Deleção de Genes , Fator Estimulador de Colônias de Granulócitos/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Recidiva Local de Neoplasia/imunologia , Resultado do Tratamento
7.
Med Sci Monit ; 24: 3764-3771, 2018 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-29864766

RESUMO

BACKGROUND Radioresistance restricts the application of radiotherapy in human osteosarcoma (OS). This study investigated the molecular mechanism of radioresistance in OS, which may provide clues to finding ideal targets for genetic therapy. MATERIAL AND METHODS The human OS cell line MG63 was employed as parent cells. After repeat low-dose X-ray irradiation of MG63, the radioresistant OS cell line MG63R was produced. Colony formation assay was used to assess the radioresistance. Cell viability was evaluated by CCK-8 assay. Flow cytometry was used to detect cell apoptosis, and wound healing assay was used to evaluate invasive capacity. The nuclear translocation was evaluated by fluorescent immunohistochemistry. Protein expression levels were assessed by Western blotting. Specific siRNA against Shh was used to silence Shh. RESULTS More survival colony formation, elevated cell viability, less cell apoptosis, and increased wound closure were found in MG63R than in MG63 cells exposed to irradiation. The nuclear translocation of Gli, expression levels of Shh, Smo, Ptch1, Bcl2, active MMP2, and active MMP9 were increased in MG63R cells compared with MG63 cells. Transfection of Shh-siRNA suppressed expression levels of Shh, Smo, Ptch1, Bcl2, active MMP2, and active MMP9, as well as the nuclear translocation of Gli in MG63R cells. The cell viability, survival colony formation, and wound closure were impaired, whereas cell apoptosis was increased, in siRNA-transfected MG63R cells than in control MG63R cells exposed to irradiation. CONCLUSIONS Activation of Shh signaling was involved in radioresistance of OS cells. Blocking this signaling can impair the radioresistance capacity of OS cells.


Assuntos
Proteínas Hedgehog/genética , Proteínas Hedgehog/efeitos da radiação , Tolerância a Radiação/genética , Apoptose/fisiologia , Linhagem Celular Tumoral/efeitos da radiação , Movimento Celular/genética , Movimento Celular/efeitos da radiação , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Sobrevivência Celular , Proteínas Hedgehog/fisiologia , Humanos , Invasividade Neoplásica/genética , Osteossarcoma/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/fisiologia
8.
Int J Clin Oncol ; 22(1): 59-69, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27553958

RESUMO

BACKGROUND: The Notch receptor plays an important role in various cell fate decisions during development and in cancer. We have previously reported that Notch3 is upregulated by radiation in non-small cell lung cancer (NSCLC) cell lines and that the Notch pathway inhibitor γ secretase inhibitor GSI (gamma-secretase inhibitor), when combined with radiation therapy, significantly suppressed the growth of lung cancer cells. However, little is known about the mechanism of Notch upregulation induced by radiation. Based on reports of Notch expression being activated through the hypoxia inducible factor 1 (HIF-1) under hypoxic conditions, we hypothesized that HIF-1 would be involved in radiation-induced Notch activation in NSCLC. METHODS: Changes in HIF-1 and Notch expression in two Notch expressing NSCLC cells line after radiation treatment were examined using Western blotting. Notch expression was evaluated after the suppression of HIF-1α by small interfering RNA. The cytotoxic effect of YC-1, a HIF inhibitor, GSI and radiation was examined using the MTT assay in vitro and the xenograft model. RESULT: We found radiation-induced expression of HIF-1α protein at 2-6 h after treatment and upregulated expression of Notch3 protein at 24 h after treatment under hypoxic conditions. Specific suppression of HIF-1α expression downregulated the radiation-induced Notch3 activation, suggesting that the Notch pathway is activated though HIF-1α after radiation. An antitumor effect of YC-1 was evident under hypoxic conditions only when there was simultaneous radiation treatment. GSI and YC-1 had a synergistic antitumor effect in vitro, and the combination of GSI and YC-1 showed the greatest radiosensitivity in vivo. CONCLUSION: Radiation-induced upregulation of the Notch pathway and HIF-1α protein may be potential therapeutic targets for more effective radiation therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/radioterapia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Neoplasias Pulmonares/radioterapia , Receptores Notch/antagonistas & inibidores , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/efeitos da radiação , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Indazóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos Nus , Terapia de Alvo Molecular/métodos , Oligopeptídeos/farmacologia , RNA Interferente Pequeno , Receptores Notch/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Childs Nerv Syst ; 33(3): 429-436, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27942918

RESUMO

BACKGROUND: Immunotherapy can be an effective treatment for pediatric medulloblastoma (MB) patients. However, major subpopulations do not respond to immunotherapy, due to the lack of antigenic mutations or the immune-evasive properties of MB cells. Clinical observations suggest that radiation therapy (RT) may expand the therapeutic reach of immunotherapy. The aim of the present investigation is to study the effect of low-dose X-ray radiation (LDXR, 1 Gy) on the functional immunological responses of MB cells (DAOY, D283, and D341). METHODS: Induction of MB cell death was examined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Production of reactive oxygen species (ROS) was measured by fluorescent probes. Changes in the expression of  human leukocyte antigen (HLA) molecules and caspase-3 activities during treatment were analyzed using Western blotting and caspase-3 assay. RESULTS: Western blot analysis demonstrated that LDXR upregulated the expression of HLA class I and HLA II molecules by more than 20% compared with control and high-dose (12 Gy) groups in vitro. Several of these HLA subtypes, such as MAGE C1, CD137, and ICAM-1, have demonstrated upregulation. In addition, LDXR increases ROS production in association with phosphorylation of NF-κB and cell surface expression of mAb target molecules (HER2 and VEGF). These data suggest that a combined LDXR and mAb therapy can create a synergistic effect in vitro. CONCLUSION: These results suggest that LDXR modulates HLA molecules, leading to alterations in T-cell/tumor-cell interaction and enhancement of T-cell-mediated MB cell death. Also, low-dose radiotherapy combined with monoclonal antibody therapy may one day augment the standard treatment for MB, but more investigation is needed to prove its utility as a new therapeutic combination for MB patients.


Assuntos
Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Antígenos HLA/metabolismo , Meduloblastoma/metabolismo , Radiação , Ligante 4-1BB/metabolismo , Análise de Variância , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta à Radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antígenos HLA/imunologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Meduloblastoma/patologia , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor ErbB-2/imunologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/imunologia
10.
Tumour Biol ; 37(1): 305-11, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26209294

RESUMO

Nasopharyngeal carcinoma (NPC) is primarily treated by chemoradiation. However, how to promote radiation sensitivity in NPC remains a challenge. Salinomycin is potentially useful for the treatment of cancer. This study aimed to explore the radiosensitivity of salinomycin on human nasopharyngeal carcinoma cell line CNE-2. CNE-2 were treated with salinomycin or irradiation, alone or in combination. The cytotoxicity effects of salinomycin were measured using CCK-8 assay. Clonogenic survival assay was used to evaluate the effects of salinomycin on the radiosensitivity of CNE-2. The changes of cell cycle distribution and apoptosis were assayed using flow cytometry. The expression of Caspase3/Bax/Bal-2 was detected by Western blotting. DNA damage was detected via γ-H2AX foci counting. The results showed that salinomycin induced apoptosis and G2/M arrest, increased Bax and cleaved Caspase3, decreased Bcl-2 expression, and increased the formation of γ-H2AX nuclear foci. These data suggest that salinomycin may be a radiosensitizer for NPC radiotherapy.


Assuntos
Neoplasias Nasofaríngeas/patologia , Piranos/química , Radiossensibilizantes/química , Apoptose , Carcinoma , Caspase 3/metabolismo , Ciclo Celular , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Separação Celular , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Histonas/metabolismo , Humanos , Carcinoma Nasofaríngeo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Tolerância a Radiação , Proteína X Associada a bcl-2/metabolismo
11.
J Neurooncol ; 130(1): 89-98, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27510952

RESUMO

Even with aggressive treatment involving radiation therapy plus temozolomide (TMZ), the prognosis for glioblastoma remains poor. We investigated the potential for targeting signal transducer and activator of transcription-3 (STAT3) to improve the therapeutic outcome in glioblastoma. We evaluated the preclinical potential of a STAT3 inhibitor, Cpd188, combined with temozolomide and radiation using in vitro assays with two established glioblastoma cell lines (U251 and U87) and two patient-derived glioblastoma cell lines (GBL12 and GBL28) as well as in vivo studies with nude mice bearing intracranial U251 xenografts. Cpd188 potentiated the radiosensitizing effect of TMZ in U251 cells, which have high p-STAT3 expression levels. The enhanced radiosensitizing effects of TMZ were associated with impaired DNA damage repair, apoptosis and reversion of the epithelial-mesenchymal transition (EMT). Cpd188 delayed in vivo tumor growth alone and in combination with radiation and TMZ. We also confirmed the radiosensitizing effect of Cpd188 in GBL28 cells, which were originated from a patient with a high level of STAT3 expression and unmethylated MGMT. Targeting STAT3 using Cpd188 could be a viable therapeutic approach for improving the outcome of current standard therapy in glioblastoma patients with high p-STAT3 expression.


Assuntos
Dacarbazina/análogos & derivados , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma , Radiossensibilizantes/farmacologia , Fator de Transcrição STAT3/metabolismo , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Ácido Benzoico/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Metilases de Modificação do DNA/metabolismo , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/farmacologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Radiação , Fator de Transcrição STAT3/antagonistas & inibidores , Sulfonamidas/farmacologia , Temozolomida , Proteínas Supressoras de Tumor/metabolismo , Cicatrização/efeitos dos fármacos , para-Aminobenzoatos/farmacologia
12.
An Acad Bras Cienc ; 88(1): 127-36, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26959322

RESUMO

Ionizing radiation induces DNA double strand breaks (DSBs) that trigger phosphorylation of the histone protein H2AX (γH2AX). Immunofluorescent staining visualizes formation of γH2AX foci, allowing their quantification. This method, as opposed to Western blot assay and Flow cytometry, provides more accurate analysis, by showing exact position and intensity of fluorescent signal in each single cell. In practice there are problems in quantification of γH2AX. This paper is based on two issues: the determination of which technique should be applied concerning the radiation dose, and how to analyze fluorescent microscopy images obtained by different microscopes. HTB140 melanoma cells were exposed to γ-rays, in the dose range from 1 to 16 Gy. Radiation effects on the DNA level were analyzed at different time intervals after irradiation by Western blot analysis and immunofluorescence microscopy. Immunochemically stained cells were visualized with two types of microscopes: AxioVision (Zeiss, Germany) microscope, comprising an ApoTome software, and AxioImagerA1 microscope (Zeiss, Germany). Obtained results show that the level of γH2AX is time and dose dependent. Immunofluorescence microscopy provided better detection of DSBs for lower irradiation doses, while Western blot analysis was more reliable for higher irradiation doses. AxioVision microscope containing ApoTome software was more suitable for the detection of γH2AX foci.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos da radiação , Relação Dose-Resposta à Radiação , Histonas/efeitos da radiação , Melanoma/radioterapia , Radiometria/métodos , Western Blotting , Linhagem Celular Tumoral/efeitos da radiação , Humanos , Microscopia de Fluorescência , Fosforilação
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 41(1): 9-18, 2016 Jan.
Artigo em Zh | MEDLINE | ID: mdl-26819419

RESUMO

OBJECTIVE: To explore the effects of 3-methyladenine (3-MA, an autophagy inhibitor) on sensitivities of nasopharyngeal carcinoma cells to radiotherapy and chemotherapy and the underlying mechanisms.
 METHODS: Cell proliferation was examined by MTT and colony formation assay, while cell apoptosis was evaluated by annexin V/PI double staining and 2-(4-Amidinophenyl)-6-indolecarbamidine dihydrochloride (DAPI) staining. Mitochondrial membrane potential was measured by commercial kit (JC-1). The expression of endoplasmic reticulum stress (ERS)-related protein, glucose-regulated protein 78 (GRP78) and autophagy-related protein beclin1, microtubule-associated protein 1 light chain 3 (LC3) were examined by Western blot.
 RESULTS: Cisplatin (DDP), ionizing radiation (IR) or tunicamycin (TM) treatment obviously inhibited the proliferation of HONE-1 cells in a concentration-dependent and time-dependent manner. Compared with control group, pretreatment with 1 mmol/L of 3-MA significantly reduced cell viability and enhanced the apoptosis in the DDP (6.00 µmol/L), 4.00 Gy IR or TM (1.00 µmol/L) groups. There was no significant difference in the apoptosis between the DDP (5.8%) and 4Gy IR (6.7%) groups. Compared with the control group, protein levels of GRP78, beclin1 and lipid-conjugated membrane-bound form (LC3-II) were significantly increased after the treatment of DDP, 4.00 Gy IR or TM, which were inhibited by pretreatment of 3-MA.
 CONCLUSION: 3-MA can sensitize HONE-1 cells to chemotherapy and radiotherapy, which is related to prevention of endoplasmic reticulum stress-induced autophagy in nasopharyngeal carcinoma cells.


Assuntos
Adenina/análogos & derivados , Autofagia , Neoplasias Nasofaríngeas/patologia , Radiossensibilizantes/farmacologia , Adenina/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Carcinoma , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Proliferação de Células , Sobrevivência Celular , Cisplatino/farmacologia , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Humanos , Potencial da Membrana Mitocondrial , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Carcinoma Nasofaríngeo , Radiação Ionizante , Tunicamicina/farmacologia
14.
Biochem Biophys Res Commun ; 456(1): 262-8, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25446081

RESUMO

Radiotherapy of is well established and frequently utilized in prostate cancer (PCa) patients. However, recurrence following therapy and distant metastases are commonly encountered problems. Previous studies underline that, in addition to its therapeutic effects, ionizing radiation (IR) increases the vascularity and invasiveness of surviving radioresistant cancer cells. This invasive phenotype of radioresistant cells is an upshot of IR-induced pro-survival and mitogenic signaling in cancer as well as endothelial cells. Here, we demonstrate that a plant flavonoid, silibinin can radiosensitize endothelial cells by inhibiting expression of pro-angiogenic factors. Combining silibinin with IR not only strongly down-regulated endothelial cell proliferation, clonogenicity and tube formation ability rather it strongly (p<0.001) reduced migratory and invasive properties of PCa cells which were otherwise marginally affected by IR treatment alone. Most of the pro-angiogenic (VEGF, iNOS), migratory (MMP-2) and EMT promoting proteins (uPA, vimentin, N-cadherin) were up-regulated by IR in PCa cells. Interestingly, all of these invasive and EMT promoting actions of IR were markedly decreased by silibinin. Further, we found that potentiated effect was an end result of attenuation of IR-activated mitogenic and pro-survival signaling, including Akt, Erk1/2 and STAT-3, by silibinin.


Assuntos
Indutores da Angiogênese/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias da Próstata/patologia , Radiação Ionizante , Silimarina/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Movimento Celular , Proliferação de Células , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos da radiação , Humanos , Masculino , Recidiva Local de Neoplasia , Fenótipo , Silibina , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
15.
BMC Cancer ; 15: 466, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26059545

RESUMO

BACKGROUND: Radiation resistance presents a challenge to the effective treatment of cancer. If therapeutic compounds were capable of resensitizing resistant tumours then a concurrent chemo-radiation treatment could be used to overcome radiation resistance. METHODS: We have developed a phenotypic assay to investigate the response of radiation resistant breast cancer cells grown in 3D-microtissue spheroids to combinations of radiation and established chemotherapeutic drugs. The effects were quantified by real time high content imaging of GFP detection area over 14 days. Ten established chemotherapeutic drugs were tested for their ability to enhance the effects of radiation. RESULTS: Of ten analysed chemotherapeutics, vinblastine was the most effective compound, with docetaxel and doxorubicine being less effective in combination with radiation. To investigate the response in a model closer to the in vivo situation we investigated the response of heterotypic 3D microtissues containing both fibroblasts and breast cancer cells. Drug treatment of these heterotypic 3D cultures confirmed treatment with radiation plus vinblastine to be additive in causing breast cancer growth inhibition. We have validated the screen by comparing radiation sensitizing effects of known chemotherapeutic agents. In both monotypic and heterotypic models the concurrent treatment of vinblastine and radiation proved more effective inhibitors of mammary cancer cell growth. The effective concentration range of both vinblastine and radiation are within the range used in treatment, suggesting the 3D model will offer a highly relevant screen for novel compounds. CONCLUSIONS: For the first time comfortable 3D cell-based phenotypic assay is available, that allows high throughput screening of compounds with radiation therapy modulating capacity, opening the field to drug discovery.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Técnicas de Cultura de Células/métodos , Tolerância a Radiação/efeitos dos fármacos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Docetaxel , Doxorrubicina/administração & dosagem , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Humanos , Taxoides/administração & dosagem , Vimblastina/administração & dosagem
16.
Clin Lab ; 61(10): 1365-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26642696

RESUMO

BACKGROUND: Osteosarcoma is the most prevalent malignant neoplasm in children and young adults with a very high propensity for local invasion and early systemic metastases. Radiotherapy has been widely used in metastatic and recurrent osteosarcoma, particularly with chemoresistance. METHODS: To determine whether autophagy is induced by radiation therapy and contributes to cell death of osteosarcoma, we investigated the influence of autophagy blockage on the radiosensitivity of osteosarcoma MG-63 cells in vitro. Firstly, autophagy in the MG-63 osteosarcoma cells after radiation treatment was determined by quantitative GFP-LC3 analysis and autophagy-related molecules analysis by western blotting. Then the viability and death of cells post-blockage of autophagy was determined by MTT assay and flow cytometry. RESULTS: It was demonstrated that autophagy was involved in MG-63 cells subject to radiation. Significantly up-regulated autophagic vesicles in MG-63 cells were subject to radiation. The transformation of LC-3 I to LC-3 II and the expression of autophagy-associated molecules were promoted in the radiation-treated MG-63 cells. Moreover, autophagy could ameliorate the cell viability post radiation. On the other hand, the chemical blockage of autophagy by 3MA not only could downregulate the level of autophagy, but also could reduce cell viability and accelerate apoptosis in the radiation-treated MG-63 cells. CONCLUSIONS: Autophagy was involved in the radiation treatment of MG-63 osteosarcoma cells, and autophagy blockage enhances the radiosensitivity of the osteosarcoma cell line MG-63 in vitro.


Assuntos
Autofagia , Osteossarcoma/metabolismo , Osteossarcoma/radioterapia , Tolerância a Radiação , Apoptose , Proteína 5 Relacionada à Autofagia , Linhagem Celular Tumoral/efeitos da radiação , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Metástase Neoplásica , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Radioterapia/métodos , Transdução de Sinais , Regulação para Cima
17.
Med Sci Monit ; 21: 1297-303, 2015 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-25943633

RESUMO

BACKGROUND: MiR-27a is significantly overexpressed in triple-negative breast cancer (TNBC). However, the exact biological function of MiR-27a in TNBC is not fully understood. In this study, we verified miR-27a expression in TNBC cells and explored how its overexpression modulates radiosensitivity of the cells. MATERIAL/METHODS: qRT-PCR analysis was performed to study miR-27a expression in TNBC lines MDA-MB-435 and MDA-MB-231 and in normal human breast epithelial cell line MCF10A. Dual luciferase assay was performed to verify a putative downstream target of miR-27a, CDC27. CCK-8 assay was used to assess the influence of miR-27a-CDC27 axis on cell proliferation under irradiation (IR) treatment. RESULTS: We confirmed significantly higher miR-27a expression in 2 TNBC cell lines--MDA-MB-435 and MDA-MB-231--than in human breast epithelial cell line MCF10A. miR-27a could modulate proliferation and radiosensitivity of TNBC cells. CDC-27 is a direct target of miR-27a and its downregulation conferred increased radioresistance of the cells. CONCLUSIONS: The miR-27a-CDC27 axis might play an important role in modulating response to radiotherapy in TNBC cells. Testing miR-27a expression might be a useful way to identify a subgroup of patients who will benefit from an IR-based therapeutic approach.


Assuntos
Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/antagonistas & inibidores , MicroRNAs/fisiologia , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/radioterapia , Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/biossíntese , Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/genética , Subunidade Apc3 do Ciclossomo-Complexo Promotor de Anáfase/fisiologia , Sítios de Ligação , Mama/citologia , Linhagem Celular Tumoral/efeitos da radiação , Células Cultivadas , Sequência Conservada , Regulação para Baixo , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Terapia de Alvo Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Oligonucleotídeos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Tolerância a Radiação/genética , Proteínas Recombinantes de Fusão/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Ensaio Tumoral de Célula-Tronco
18.
Mol Cell Proteomics ; 12(2): 485-98, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23204318

RESUMO

Tumor hypoxia induces cancer cell angiogenesis, invasiveness, treatment resistance, and contributes to poor clinical outcome. However, the molecular mechanism by which tumor hypoxia exerts a coordinated effect on different molecular pathways to enhance tumor growth and survival and lead to poor clinical outcome is not fully understood. In this study, we attempt to elucidate the global protein expression and functional changes in A431 epithelial carcinoma cells induced by hypoxia and reoxygenation using iTRAQ quantitative proteomics and biochemical functional assays. Quantitative proteomics results showed that 4316 proteins were quantified with FDR<1%, in which over 1200 proteins were modulated >1.2 fold, and DNA repair, glycolysis, integrin, glycoprotein turnover, and STAT1 pathways were perturbed by hypoxia and reoxygenation-induced oxidative stress. For the first time, hypoxia was shown to up-regulate the nonhomologous end-joining pathway, which plays a central role in DNA repair of irradiated cells, thereby potentially contributing to the radioresistance of hypoxic A431 cells. The up-regulation of Ku70/Ku80 dimer, a key molecular complex in the nonhomologous end-joining pathway, was confirmed by Western blot and liquid chromatography/tandem mass spectrometry-MRM methods. Functional studies confirmed that up-regulation of glycolysis, integrin, glycoprotein synthesis, and down-regulation of STAT1 pathways during hypoxia enhanced metastastic activity of A431 cells. Migration of A431 cells was dramatically repressed by glycolysis inhibitor (2-Deoxy-d-glucose), glycoprotein synthesis inhibitor (1-Deoxynojirimycin Hydrochloride), and STAT1α overexpression that enhanced the integrin-mediated cell adhesion. These results revealed that hypoxia induced several biological processes involved in tumor migration and radioresistance and provided potential new targets for tumor therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Tolerância a Radiação , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Adesão Celular , Hipóxia Celular , Linhagem Celular Tumoral/efeitos da radiação , Movimento Celular , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Raios gama , Humanos , Integrinas/genética , Integrinas/metabolismo , Autoantígeno Ku , Neoplasias/radioterapia , Estresse Oxidativo , Multimerização Proteica , Inibidores da Síntese de Proteínas/farmacologia , Proteômica , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
19.
BMC Public Health ; 15: 764, 2015 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-26253141

RESUMO

BACKGROUND: The increase in mobile phone use has generated concerns about possible risks to human health, especially the development of brain tumors. Whether tumor patients should continue to use mobile telephones has remained unclear because of a paucity of information. Herein, we investigated whether electromagnetic fields from mobile phones could alter the biological features of human tumor cells and act as a tumor-promoting agent. METHODS: Human glioblastoma cell lines, U251-MG and U87-MG, were exposed to 1950-MHz time division-synchronous code division multiple access (TD-SCDMA) at a specific absorption rate (maximum SAR = 5.0 W/kg) for 12, 24, and 48 h. Cell morphologies and ultra-structures were observed by microscopy and the rates of apoptosis and cell cycle progression were monitored by flow cytometry. Additionally, cell growth was determined using the CKK-8 assay, and the expression levels of tumor and apoptosis-related genes and proteins were analyzed by real-time PCR and western blotting, respectively. Tumor formation and invasiveness were measured using a tumorigenicity assay in vivo and migration assays in vitro. RESULTS: No significant differences in either biological features or tumor formation ability were observed between unexposed and exposed glioblastoma cells. Our data showed that exposure to 1950-MHz TD-SCDMA electromagnetic fields for up to 48 h did not act as a cytotoxic or tumor-promoting agent to affect the proliferation or gene expression profile of glioblastoma cells. CONCLUSIONS: Our findings implied that exposing brain tumor cells in vitro for up to 48 h to 1950-MHz continuous TD-SCDMA electromagnetic fields did not elicit a general cell stress response.


Assuntos
Apoptose/efeitos da radiação , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos da radiação , Telefone Celular , Campos Eletromagnéticos/efeitos adversos , Glioblastoma/patologia , Neoplasias Encefálicas/química , Neoplasias Encefálicas/prevenção & controle , Linhagem Celular Tumoral/efeitos da radiação , Fenômenos Fisiológicos Celulares/efeitos da radiação , Glioblastoma/química , Glioblastoma/prevenção & controle , Proteínas de Choque Térmico/análise , Humanos , Doses de Radiação
20.
Gen Physiol Biophys ; 34(1): 13-21, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25395599

RESUMO

Photodynamic therapy (PDT) is a method to treat cancers using photosensitizer and light. PDT has been tried for several tumors. However, the clinical applications are limited by the toxicity of photosensitizer and narrow effect. Sulforaphane (SFN) is a material of isothiocyanate group and known to have anticancer effect. We evaluated the cytotoxic effect of PDT combined with SFN on human head and neck cancer cells. We measured the cell viability, extent of apoptosis and necrosis, reactive oxygen species (ROS) generation and caspase activation. Cell viability was decreased significantly by combination treatment. Cellular apoptosis and necrosis were increased in combination treatment compared to SFN or PDT. ROS generation was also higher in combination treatment than single treatment. In combination treatment group, apoptosis and necrosis were decreased by administration of sodium azide (SA) which is scavenger of ROS. Increased caspase activation in combination treatment was also inhibited by SA. Combination of PDT and SFN led to enhanced cytotoxic effect on head and neck cancer cells. Combination treatment promoted the ROS generation, which induced cell death through activation of caspase pathway.


Assuntos
Neoplasias de Cabeça e Pescoço/patologia , Isotiocianatos/farmacologia , Fotoquimioterapia/métodos , Antineoplásicos/farmacologia , Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos da radiação , Separação Celular , Sobrevivência Celular , Citometria de Fluxo , Humanos , Microscopia Confocal , Necrose , Espécies Reativas de Oxigênio/metabolismo , Azida Sódica/farmacologia , Sulfóxidos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa