Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(11): e44367, 2012.
Article in English | MEDLINE | ID: mdl-23185232

ABSTRACT

Poor local control and tumor escape are of major concern in head-and-neck cancers treated by conventional radiotherapy or hadrontherapy. Reduced glutathione (GSH) is suspected of playing an important role in mechanisms leading to radioresistance, and its depletion should enable oxidative stress insult, thereby modifying the nature of DNA lesions and the subsequent chromosomal changes that potentially lead to tumor escape.This study aimed to highlight the impact of a GSH-depletion strategy (dimethylfumarate, and L-buthionine sulfoximine association) combined with carbon ion or X-ray irradiation on types of DNA lesions (sparse or clustered) and the subsequent transmission of chromosomal changes to the progeny in a radioresistant cell line (SQ20B) expressing a high endogenous GSH content. Results are compared with those of a radiosensitive cell line (SCC61) displaying a low endogenous GSH level. DNA damage measurements (γH2AX/comet assay) demonstrated that a transient GSH depletion in resistant SQ20B cells potentiated the effects of irradiation by initially increasing sparse DNA breaks and oxidative lesions after X-ray irradiation, while carbon ion irradiation enhanced the complexity of clustered oxidative damage. Moreover, residual DNA double-strand breaks were measured whatever the radiation qualities. The nature of the initial DNA lesions and amount of residual DNA damage were similar to those observed in sensitive SCC61 cells after both types of irradiation. Misrepaired or unrepaired lesions may lead to chromosomal changes, estimated in cell progeny by the cytome assay. Both types of irradiation induced aberrations in nondepleted resistant SQ20B and sensitive SCC61 cells. The GSH-depletion strategy prevented the transmission of aberrations (complex rearrangements and chromosome break or loss) in radioresistant SQ20B only when associated with carbon ion irradiation. A GSH-depleting strategy combined with hadrontherapy may thus have considerable advantage in the care of patients, by minimizing genomic instability and improving the local control.


Subject(s)
Carbon/chemistry , Chromosomes, Human/metabolism , DNA Damage , Glutathione/deficiency , Neoplasms/metabolism , Neoplasms/pathology , Radiation , Buthionine Sulfoximine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chromatography, High Pressure Liquid , Clone Cells , Cluster Analysis , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Breaks, Single-Stranded/drug effects , DNA Breaks, Single-Stranded/radiation effects , Dimethyl Fumarate , Fumarates/pharmacology , Gene Rearrangement/drug effects , Gene Rearrangement/radiation effects , Glutathione/metabolism , Histones/metabolism , Humans , Ions , Kinetics , Micronucleus Tests , X-Rays
2.
PLoS One ; 6(1): e14558, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21283807

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is an aggressive and recurrent malignancy owing to intrinsic radioresistance and lack of induction of apoptosis. The major focus of this work was to design a transient glutathione depleting strategy during the course of irradiation of HNSCC in order to overcome their radioresistance associated with redox adaptation. METHODOLOGY/PRINCIPAL FINDINGS: Treatment of SQ20B cells with dimethylfumarate (DMF), a GSH-depleting agent, and L-Buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis 4 h before a 10 Gy irradiation led to the lowering of the endogenous GSH content to less than 10% of that in control cells and to the triggering of radiation-induced apoptotic cell death. The sequence of biochemical events after GSH depletion and irradiation included ASK-1 followed by JNK activation which resulted in the triggering of the intrinsic apoptotic pathway through Bax translocation to mitochondria. CONCLUSIONS: This transient GSH depletion also triggered radiation-induced cell death in SQ20B stem cells, a key event to overcome locoregional recurrence of HNSCC. Finally, our in vivo data highlight the relevance for further clinical trials of endogenous redox modulation to enhance the cytotoxic effects of radiotherapy.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Neoplastic Stem Cells , Adaptation, Physiological , Apoptosis/drug effects , Apoptosis/radiation effects , Buffers , Buthionine Sulfoximine/pharmacology , Carcinoma/pathology , Carcinoma/therapy , Cell Line, Tumor , Dimethyl Fumarate , Fumarates/pharmacology , Glutathione/antagonists & inhibitors , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/therapy , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Neoplasms, Squamous Cell/pathology , Neoplasms, Squamous Cell/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Oxidation-Reduction , Squamous Cell Carcinoma of Head and Neck , bcl-2-Associated X Protein/metabolism
3.
Int J Radiat Oncol Biol Phys ; 75(4): 1247-53, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19857788

ABSTRACT

PURPOSE: Free radicals are believed to play an active role in the bystander response. This study investigated their origin as well as their temporal and spatial impacts in the bystander effect. METHODS AND MATERIALS: We employed a precise alpha-particle microbeam to target a small fraction of subconfluent osteoblastic cells (MC3T3-E1). gammaH2AX-53BP1 foci, oxidative metabolism changes, and micronuclei induction in targeted and bystander cells were assessed. RESULTS: Cellular membranes and mitochondria were identified as two distinct reactive oxygen species producers. The global oxidative stress observed after irradiation was significantly attenuated after cells were treated with filipin, evidence for the primal role of membrane in the bystander effect. To determine the membrane's impact at a cellular level, micronuclei yield was measured when various fractions of the cell population were individually targeted while the dose per cell remained constant. Induction of micronuclei increased in bystander cells as well as in targeted cells and was attenuated by filipin treatment, demonstrating a role for bystander signals between irradiated cells in an autocrine/paracrine manner. CONCLUSIONS: A complex interaction of direct irradiation and bystander signals leads to a membrane-dependent amplification of cell responses that could influence therapeutic outcomes in tissues exposed to low doses or to environmental exposure.


Subject(s)
Bystander Effect/physiology , Cell Membrane/metabolism , Mitochondria/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , 3T3 Cells , Alpha Particles/therapeutic use , Animals , Bystander Effect/drug effects , DNA Breaks, Double-Stranded , Filipin/pharmacology , Glutathione/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Micronucleus Tests , Osteoblasts/metabolism , Osteoblasts/radiation effects , Osteoblasts/ultrastructure , Oxidative Stress/drug effects , Tumor Suppressor p53-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...