Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 3853, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594122

RESUMEN

This work combines single cell Raman spectroscopy (RS) with group and basis restricted non-negative matrix factorisation (GBR-NMF) to identify individual biochemical changes associated with radiation exposure in three human cancer cell lines. The cell lines analysed were derived from lung (H460), breast (MCF7) and prostate (LNCaP) tissue and are known to display varying degrees of radio sensitivity due to the inherent properties of each cell type. The GBR-NMF approach involves the deconstruction of Raman spectra into component biochemical bases using a library of Raman spectra of known biochemicals present in the cells. Subsequently, scores are obtained on each of these bases which can be directly correlated with the contribution of each chemical to the overall Raman spectrum. We validated GBR-NMF through the correlation of GBR-NMF-derived glycogen scores with scores that were previously observed using principal component analysis (PCA). Phosphatidylcholine, glucose, arginine and asparagine showed a distinct differential score pattern between radio-resistant and radio-sensitive cell types. In summary, the GBR-NMF approach allows for the monitoring of individual biochemical radiation-response dynamics previously unattainable with more traditional PCA-based approaches.


Asunto(s)
Células MCF-7/metabolismo , Células MCF-7/efectos de la radiación , Modelos Biológicos , Glucógeno/metabolismo , Humanos , Espectrometría Raman , Aprendizaje Automático Supervisado
2.
Drug Deliv ; 27(1): 791-804, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32420760

RESUMEN

The combined chemotherapy and photodynamic therapy have significant advantages for cancer treatments, which have higher therapeutic effects compared with other medicines. Herein, we focused on the synthesis of carbon quantum dot (CQD) based nanocarrier system. CQD and 5-aminolevulinic acid (5-ALA) were conjugated with mono-(5-BOC-protected-glutamine-6-deoxy) ß-cyclodextrin (CQD-Glu-ß-CD) moiety, and finally, the anticancer chemotherapy doxorubicin (DOX) drug was loaded in the 5-ALA-CQD-Glu-ß-CD system. The stepwise physicochemical changes for the preparation of the DOX loaded 5-ALA-CQD-Glu-ß-CD system were investigated by Fourier transform infrared (FT-IR) spectroscopy, X-ray diffraction (XRD), transmission electron microscopy (TEM), atomic force microscopy (AFM), and Raman fluorescence spectroscopy. The encapsulation efficiency of DOX in 5-ALA-CQD-Glu-ß-CD was observed at ∼83.0%, and the loading capacity of DOX is ∼20.37%. The in vitro releasing of DOX and 5-ALA was observed through the UV-vis spectroscopy by the λmax value of 487 nm and 253 nm, respectively. By the investigation against the breast MCF-7 cancer cells, the high cytotoxicity and morphological changes of cancer cells were observed by the treating of DOX/5-ALA-CQD-Glu-ß-CD. The generation of reactive oxygen species (ROS) upon 635 nm (25 mW cm-2) for 15 min laser irradiation-induced improved the therapeutic effects. In vitro cellular uptake studies recommend the synthesized DOX/5-ALA-CQD-Glu-ß-CD nanocarrier could significantly enhance the cell apoptosis and assist in the MCF-7 cell damages. The result suggests a multifunctional therapeutic system for chemo/photodynamic synergistic effects on cancer therapy.


Asunto(s)
Ácido Aminolevulínico , Antineoplásicos , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/farmacología , Fotoquimioterapia/métodos , beta-Ciclodextrinas , Ácido Aminolevulínico/química , Ácido Aminolevulínico/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carbono/química , Terapia Combinada , Sistemas de Liberación de Medicamentos/métodos , Humanos , Terapia por Láser/métodos , Células MCF-7/efectos de los fármacos , Células MCF-7/efectos de la radiación , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Puntos Cuánticos/química , Puntos Cuánticos/uso terapéutico , Resultado del Tratamiento , beta-Ciclodextrinas/química , beta-Ciclodextrinas/farmacología
3.
Radiat Prot Dosimetry ; 186(2-3): 155-158, 2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-31803908

RESUMEN

A comparison between breast cancer cell line MCF7 and human adipose-derived stem cells (ADSC) after irradiation by the same doses of megavoltage X-rays was performed. The cell growth, the induction of apoptosis and the expression of selected genes were analyzed. Irradiated MCF7 related to its control sample grows slower than ADSC and it undergoes apoptosis in much higher levels than ADSC. This was confirmed by real-time polymerase chain reaction as well, where the expression of apoptotic genes was found to be considerably higher for MCF7 than for ADSC. From the results of this project, it could be stated that MCF7 is more radiosensitive than ADSC.


Asunto(s)
Adipocitos/efectos de la radiación , Células MCF-7/efectos de la radiación , Tolerancia a Radiación , Células Madre/efectos de la radiación , Apoptosis/efectos de la radiación , Proliferación Celular/efectos de la radiación , Humanos
4.
Ginekol Pol ; 89(4): 175-182, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29781071

RESUMEN

OBJECTIVES: Intraoperative radiotherapy (IORT) relates to irradiation of diseased tissue during the surgery within the tumor bed. The reason for this process is based on the fact that the increase in the radiation dose increases local tumor control. It was shown that postoperative fluids obtained from patients after breast cancer conserving surgery, stimulated motility and invasiveness of tumor cells in vitro. The results obtained from TARGIT clinical trial demonstrated that IORT significantly inhibits the stimulatory effect of wound fluids on tumor cells in vitro. We therefore speculated that wound fluids collected from patients after IORT treatment may induce the apoptosis in breast cancer cell lines and it may be a reason for their lower proliferation rate and potential to metastasis. MATERIAL AND METHODS: Breast cancer MCF7 cell line was incubated with wound fluids collected from patients after conserving breast cancer surgery or surgery followed by IORT for 4 days. Then the expression of markers associated with extrinsic or intrinsic apoptosis pathway was established. RESULTS: Our results clearly indicate activation of extrinsic apoptosis pathway by wound fluids collected from patients after IORT treatment. No changes in apoptotic markers were seen in cells treated with wound fluids collected from patients after the surgery alone. CONCLUSIONS: Thus we confirmed that wound fluids collected from patients after IORT treatment may induce the apoptosis in breast cancer cell lines and it may be a reason for their lower proliferation rate and invasiveness of tumor cells in vitro.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/radioterapia , Proliferación Celular/efectos de la radiación , Quimioradioterapia Adyuvante/efectos adversos , Células MCF-7/efectos de la radiación , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Periodo Intraoperatorio , Persona de Mediana Edad
5.
J Med Chem ; 59(7): 3003-17, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26978566

RESUMEN

Amifostine protects normal cells from DNA damage induction by ionizing radiation or chemotherapeutics, whereas cancer cells typically remain uninfluenced. While confirming this phenomenon, we have revealed by comet assay and currently the most sensitive method of DNA double strand break (DSB) quantification (based on γH2AX/53BP1 high-resolution immunofluorescence microscopy) that amifostine treatment supports DSB repair in γ-irradiated normal NHDF fibroblasts but alters it in MCF7 carcinoma cells. These effects follow from the significantly lower activity of alkaline phosphatase measured in MCF7 cells and their supernatants as compared with NHDF fibroblasts. Liquid chromatography-mass spectrometry confirmed that the amifostine conversion to WR-1065 was significantly more intensive in normal NHDF cells than in tumor MCF cells. In conclusion, due to common differences between normal and cancer cells in their abilities to convert amifostine to its active metabolite WR-1065, amifostine may not only protect in multiple ways normal cells from radiation-induced DNA damage but also make cancer cells suffer from DSB repair alteration.


Asunto(s)
Amifostina/farmacología , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Protectores contra Radiación/farmacología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Amifostina/farmacocinética , Ensayo Cometa , Roturas del ADN de Doble Cadena/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Rayos gamma , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células MCF-7/efectos de los fármacos , Células MCF-7/efectos de la radiación , Mercaptoetilaminas/farmacocinética , Microscopía Fluorescente/métodos , Proteína 1 de Unión al Supresor Tumoral P53
6.
DNA Cell Biol ; 35(3): 140-5, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26717101

RESUMEN

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.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Rayos Ultravioleta/efectos adversos , Citoplasma/metabolismo , Citoplasma/efectos de la radiación , Citoesqueleto/metabolismo , Enzima Desubiquitinante CYLD , Células HeLa/efectos de la radiación , Humanos , Células MCF-7/efectos de la radiación , Microtúbulos/metabolismo , Microtúbulos/efectos de la radiación , Procesamiento Proteico-Postraduccional/efectos de la radiación , Transporte de Proteínas/efectos de la radiación
7.
Acta Biochim Pol ; 62(2): 173-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26015993

RESUMEN

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.


Asunto(s)
Heterocromatina/metabolismo , Histonas/metabolismo , Radiación Ionizante , Daño del ADN/fisiología , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células HeLa/metabolismo , Células HeLa/efectos de la radiación , Heterocromatina/efectos de la radiación , Humanos , Lisina/metabolismo , Células MCF-7/metabolismo , Células MCF-7/efectos de la radiación , Metilación , Tolerancia a Radiación , Proteína p53 Supresora de Tumor/metabolismo
8.
Int J Nanomedicine ; 10: 2065-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25844037

RESUMEN

Cancer ranks among the leading causes of human mortality. Cancer becomes intractable when it spreads from the primary tumor site to various organs (such as bone, lung, liver, and then brain). Unlike solid tumor cells, cancer stem cells and metastatic cancer cells grow in a non-attached (suspension) form when moving from their source to other locations in the body. Due to the non-attached growth nature, metastasis is often first detected in the circulatory systems, for instance in a lymph node near the primary tumor. Cancer research over the past several decades has primarily focused on treating solid tumors, but targeted therapy to treat cancer stem cells and cancer metastasis has yet to be developed. Because cancers undergo faster metabolism and consume more glucose than normal cells, glucose was chosen in this study as a reagent to target cancer cells. In particular, by covalently binding gold nanoparticles (GNPs) with thio-PEG (polyethylene glycol) and thio-glucose, the resulting functionalized GNPs (Glu-GNPs) were created for targeted treatment of cancer metastasis and cancer stem cells. Suspension cancer cell THP-1 (human monocytic cell line derived from acute monocytic leukemia patients) was selected because it has properties similar to cancer stem cells and has been used as a metastatic cancer cell model for in vitro studies. To take advantage of cancer cells' elevated glucose consumption over normal cells, different starvation periods were screened in order to achieve optimal treatment effects. Cancer cells were then fed using Glu-GNPs followed by X-ray irradiation treatment. For comparison, solid tumor MCF-7 cells (breast cancer cell line) were studied as well. Our irradiation experimental results show that Glu-GNPs are better irradiation sensitizers to treat THP-1 cells than MCF-7 cells, or Glu-GNPs enhance the cancer killing of THP-1 cells 20% more than X-ray irradiation alone and GNP treatment alone. This finding can help oncologists to design therapeutic strategies to target cancer stem cells and cancer metastasis.


Asunto(s)
Antineoplásicos/farmacología , Glucosa/química , Oro/química , Nanopartículas del Metal , Células Madre Neoplásicas/efectos de los fármacos , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/efectos de la radiación , Femenino , Oro/farmacología , Humanos , Células MCF-7/efectos de los fármacos , Células MCF-7/efectos de la radiación , Nanopartículas del Metal/química , Células Madre Neoplásicas/metabolismo , Polietilenglicoles/farmacología , Rayos X
9.
Appl Spectrosc ; 69(2): 193-204, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25588147

RESUMEN

The drive toward personalized radiation therapy (RT) has created significant interest in determining patient-specific tumor and normal tissue responses to radiation. Raman spectroscopy (RS) is a non-invasive and label-free technique that can detect radiation response through assessment of radiation-induced biochemical changes in tumor cells. In the current study, single-cell RS identified specific radiation-induced responses in four human epithelial tumor cell lines: lung (H460), breast (MCF-7, MDA-MB-231), and prostate (LNCaP), following exposure to clinical doses of radiation (2-10 Gy). At low radiation doses (2 Gy), H460 and MCF-7 cell lines showed an increase in glycogen-related spectral features, and the LNCaP cell line showed a membrane phospholipid-related radiation response. In these cell lines, only spectral information from populations receiving 10 Gy or less was required to identify radiation-related features using principal component analysis (PCA). In contrast, the MDA-MB-231 cell line showed a significant increase in protein relative to nucleic acid and lipid spectral features at doses of 6 Gy or higher, and high-dose information (30, 50 Gy) was required for PCA to identify this biological response. The biochemical nature of the radiation-related changes occurring in cells exposed to clinical doses was found to segregate by status of p53 and radiation sensitivity. Furthermore, the utility of RS to identify a biological response in human tumor cells exposed to therapeutic doses of radiation was found to be governed by the extent of the biochemical changes induced by a radiation response and is therefore cell line specific. The results of this study demonstrate the utility and effectiveness of single-cell RS to identify and measure biological responses in tumor cells exposed to standard radiotherapy doses.


Asunto(s)
Radiación Ionizante , Espectrometría Raman/métodos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Genes p53 , Humanos , Células MCF-7/efectos de la radiación , Masculino , Fosfolípidos/metabolismo , Análisis de Componente Principal , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , Dosificación Radioterapéutica , Procesamiento de Señales Asistido por Computador , Análisis de la Célula Individual/métodos , Resultado del Tratamiento
10.
Int J Oncol ; 46(3): 1051-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25501936

RESUMEN

The functions of the high mobility group box 1 (HMGB1) in tumor cells include replenishing telomeric DNA and maintaining cell immortality. There is a negative correlation between human telomerase reverse transcriptase (hTERT) and radiosensitivity in tumor cells. Our aim was to elucidate the relationship among HMGB1, telomere homeostasis and radiosensitivity in MCF-7 cells. In this study, we established stably transfected control (MCF-7-NC) and HMGB1 knockdown (MCF-7-shHMGB1) cell lines. The expression of HMGB1 mRNA and the relative telomere length were examined by real-time PCR. Radiosensitivity was detected by clonogenic assay. The protein expressions were determined by western blot analysis. The telomerase activity was detected by PCR-ELISA. Proliferation ability was examined by CCK-8 assay. Cell cycle and apoptosis were examined by flow cytometry. DNA damage foci were detected by immunofluorescence. ShRNA-mediated downregulation of HMGB1 expression increased the radiosensitivity of MCF-7 cells, and reduced the accumulation of hTERT and cyclin D1. Moreover, knockdown of HMGB1 in MCF-7 cells inhibited telomerase activity and cell proliferation, while increasing the extent of apoptosis. Downregulation of HMGB1 modulated telomere homeostasis by changing the level of telomere-binding proteins, such as TPP1 (PTOP), TRF1 and TRF2. This downregulation also inhibited the ATM and ATR signaling pathways. The current data demonstrate that knockdown of HMGB1 breaks telomere homeostasis, enhances radiosensitivity, and suppresses the repair of DNA damage in human breast cancer cells. These results suggested that HMGB1 might be a potential radiotherapy target in human breast cancer.


Asunto(s)
Neoplasias de la Mama/radioterapia , Proteína HMGB1/genética , Homeostasis del Telómero/fisiología , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclo Celular/genética , Proliferación Celular/genética , Daño del ADN/genética , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Proteína HMGB1/metabolismo , Humanos , Células MCF-7/efectos de la radiación , Tolerancia a Radiación/genética , Radiación Ionizante , Serina Proteasas/genética , Serina Proteasas/metabolismo , Complejo Shelterina , Telomerasa/genética , Telomerasa/metabolismo , Proteínas de Unión a Telómeros , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
11.
Genes Cells ; 20(3): 191-202, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25482373

RESUMEN

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.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Complejo Mediador/metabolismo , Factores de Transcripción/metabolismo , Inhibidores Enzimáticos/farmacología , Células HeLa/efectos de la radiación , Humanos , Imidazoles/farmacología , Células MCF-7/efectos de los fármacos , Células MCF-7/efectos de la radiación , Complejo Mediador/genética , Piperazinas/farmacología , Unión Proteica , Transporte de Proteínas/efectos de la radiación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Polimerasa II/metabolismo , Activación Transcripcional , Rayos Ultravioleta
12.
Radiats Biol Radioecol ; 54(3): 256-64, 2014.
Artículo en Ruso | MEDLINE | ID: mdl-25764829

RESUMEN

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.


Asunto(s)
Rayos gamma , Neoplasias Laríngeas/radioterapia , Células Madre Neoplásicas/efectos de la radiación , Tolerancia a Radiación/genética , Adulto , Anciano , Animales , Femenino , Células HeLa/efectos de la radiación , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/inmunología , Inmunofenotipificación , Neoplasias Laríngeas/patología , Células MCF-7/efectos de la radiación , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/ultraestructura
13.
Oncol Rep ; 30(5): 2119-26, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24002052

RESUMEN

Emerging evidence indicates that breast cancer-initiating cells (CICs) are relatively resistant to radiotherapy; however, the critical mechanisms determining breast CIC resistance to radiation remain elusive. In the present study, a subpopulation of cells displaying characteristics generally attributed to stem cells was identified within the breast cancer cell line MCF-7. This subpopulation displays cancer stem cell features characterized by overexpression of embryonic stem cell markers, high tumorigenic potential following transplantation into BALB/c-nu mice, self-renewal capacity and resistance to ionizing radiation (IR). Moreover, glucose­regulated protein 78KD (GRP78), which was found to play a crucial role in stem cell oncogenesis, was also shown to be overexpressed in this subpopulation. GRP78 is required for the cancer stem-like subpopulation cell resistance to IR, as knockdown of this gene augments the effects of IR, while overexpression of GRP78 increases the radiation resistance of the subpopulation to IR. These findings indicate that GRP78 acts as a potential therapeutic target aimed at tumor-generating subsets of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/radioterapia , Proteínas de Choque Térmico/genética , Tolerancia a Radiación/genética , Animales , Neoplasias de la Mama/patología , Proliferación Celular/efectos de la radiación , Transformación Celular Neoplásica , Chaperón BiP del Retículo Endoplásmico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7/efectos de la radiación , Ratones , Células Madre Neoplásicas/efectos de la radiación , Radiación Ionizante
14.
Oncogene ; 32(39): 4634-45, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23108394

RESUMEN

FOXM1 is implicated in genotoxic drug resistance but its role and mechanism of action remain unclear. Here, we establish that γH2AX foci, indicative of DNA double-strand breaks (DSBs), accumulate in a time-dependent manner in the drug-sensitive MCF-7 cells but not in the resistant counterparts in response to epirubicin. We find that FOXM1 expression is associated with epirubicin sensitivity and DSB repair. Ectopic expression of FOXM1 can increase cell viability and abrogate DSBs sustained by MCF-7 cells following epirubicin, owing to an enhancement in repair efficiency. Conversely, alkaline comet and γH2AX foci formation assays show that Foxm1-null cells are hypersensitive to DNA damage, epirubicin and γ-irradiation. Furthermore, we find that FOXM1 is required for DNA repair by homologous recombination (HR) but not non-homologous end joining (NHEJ), using HeLa cell lines harbouring an integrated direct repeat green fluorescent protein reporter for DSB repair. We also identify BRIP1 as a direct transcription target of FOXM1 by promoter analysis and chromatin-immunoprecipitation assay. In agreement, depletion of FOXM1 expression by small interfering RNA downregulates BRIP1 expression at the protein and mRNA levels in MCF-7 and the epirubicin-resistant MCF-7 Epi(R) cells. Remarkably, the requirement for FOXM1 for DSB repair can be circumvented by reintroduction of BRIP1, suggesting that BRIP1 is an important target of FOXM1 in DSB repair. Indeed, like FOXM1, BRIP1 is needed for HR. These data suggest that FOXM1 regulates BRIP1 expression to modulate epirubicin-induced DNA damage repair and drug resistance.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos/fisiología , Epirrubicina/farmacología , Factores de Transcripción Forkhead/fisiología , Proteínas de Neoplasias/fisiología , ARN Helicasas/fisiología , Reparación del ADN por Recombinación/fisiología , Animales , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Femenino , Fibroblastos , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Rayos gamma , Histonas/análisis , Humanos , Células MCF-7/efectos de los fármacos , Células MCF-7/metabolismo , Células MCF-7/efectos de la radiación , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Helicasas/biosíntesis , ARN Helicasas/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Interferente Pequeño/farmacología , Tolerancia a Radiación , Proteínas Recombinantes de Fusión/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...