Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Radiat Res ; 194(5): 511-518, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33045074

ABSTRACT

Several studies have demonstrated that mitochondria are critically involved in the pleiotropic manifestation of radiation effects. While conventional whole-cell irradiation compromises the function of mitochondria, the effects of subcellular targeted radiation are not yet fully understood. In this study, normal human diploid cells with cell-cycle indicators were irradiated using a synchrotron X-ray microbeam, and mitochondrial membrane potential was quantified by JC-1 over the 72-h period postirradiation. Cytoplasmic irradiation was observed to temporarily enlarge the mitochondrial area with high membrane potential, while the total mitochondrial area did not change significantly. Unexpectedly, cell-nucleus irradiation promoted a similar increase not only in the mitochondrial areas with high membrane potential, but also in those with low membrane potential, which gave rise to the apparent increase in the total mitochondrial area. Augmentation of the mitochondrial area with low membrane potential was predominantly observed among G1 cells, suggesting that nucleus irradiation during the G1 phase regulated the mitochondrial dynamics of the cytoplasm, presumably through DNA damage in the nucleus.


Subject(s)
Cell Nucleus/radiation effects , Fibroblasts/radiation effects , Mitochondria/radiation effects , Benzimidazoles , Carbocyanines , Cells, Cultured , Cytoplasm/radiation effects , DNA Damage , Fibroblasts/ultrastructure , Fluorescent Dyes , G1 Phase/radiation effects , Humans , Membrane Potential, Mitochondrial/radiation effects , Microscopy, Fluorescence , Mitochondrial Size/radiation effects , Synchrotrons
2.
J Biophotonics ; 12(6): e201800411, 2019 06.
Article in English | MEDLINE | ID: mdl-30701682

ABSTRACT

OBJECTIVE: Photobiomodulation (PBM) is the application of light to promote tissue healing. Current indications suggest PBM induces its beneficial effects in vivo through upregulation of mitochondrial activity. However, how mitochondrial content influences such PBM responses have yet to be evaluated. Hence, the current study assessed the biological response of cells to PBM with varying mitochondrial contents. METHODS: DNA was isolated from myoblasts and myotubes (differentiated myoblasts), and mitochondrial DNA (mtDNA) was amplified and quantified using a microplate assay. Cells were seeded in 96-wellplates, incubated overnight and subsequently irradiated using a light-emitting diode array (400, 450, 525, 660, 740, 810, 830 and white light, 24 mW/cm2 , 30-240 seconds, 0.72-5.76J/cm2 ). The effects of PBM on markers of mitochondrial activity including reactive-oxygen-species and real-time mitochondrial respiration (Seahorse XFe96) assays were assessed 8 hours post-irradiation. Datasets were analysed using general linear model followed by one-way analysis of variance (and post hoc-Tukey tests); P = 0.05). RESULTS: Myotubes exhibited mtDNA levels 86% greater than myoblasts (P < 0.001). Irradiation of myotubes at 400, 450 or 810 nm induced 53%, 29% and 47% increases (relative to non-irradiated control) in maximal respiratory rates, respectively (P < 0.001). Conversely, irradiation of myoblasts at 400 or 450 nm had no significant effect on maximal respiratory rates. CONCLUSION: This study suggests that mitochondrial content may influence cellular responses to PBM and as such explain the variability of PBM responses seen in the literature.


Subject(s)
Low-Level Light Therapy , Mitochondria/radiation effects , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Animals , Cell Line , Mice , Mitochondria/metabolism , Mitochondrial Size/radiation effects
3.
Int J Radiat Biol ; 93(8): 764-770, 2017 08.
Article in English | MEDLINE | ID: mdl-28463025

ABSTRACT

PURPOSE: Tumor resistance towards radiation has been a big obstacle in the poor prognosis of lung cancer. It has been reported that hypoxia and autophagy partly contribute to this resistance. However, there is controversy over whether autophagy plays a positive role in cancer therapy or not. We aim to find out the specific mechanism of radiation resistance. MATERIALS AND METHODS: A549 cells were treated with conditioned medium (CM) under 12 h hypoxia or normoxia before irradiation, followed by the measurement of clonogenic survival, reactive oxygen species (ROS), signal of mitochondria and autophagy flux. In some experiments, the A549 cells were respectively transfected with LC3 small interfering RNA (siRNA), or treated with Earle's Balanced Salt Solution (EBSS). RESULTS: We found that hypoxia enhanced cell radioresistance by increasing the induction of autophagy. And after hypoxia stress, the number of mitochondria was reduced but the cellular ROS level was enhanced. It was significant that autophagy may enhance cell radioresistance by reducing ROS during hypoxic treatment. CONCLUSIONS: We elucidated the possible mechanisms of autophagy in regulating cancer cell death or survival. These results supply a new opinion about the intrinsic factor of radioresistance of hypoxia tumors.


Subject(s)
Autophagy/radiation effects , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Radiation Tolerance/radiation effects , Reactive Oxygen Species/metabolism , Tumor Hypoxia/radiation effects , Cell Line, Tumor , Humans , Mitochondrial Size/radiation effects
4.
Radiat Res ; 183(6): 594-609, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25973951

ABSTRACT

Hypoxia is a major cause of radiation resistance, which may predispose to local recurrence after radiation therapy. While hypoxia increases tumor cell survival after radiation exposure because there is less oxygen to oxidize damaged DNA, it remains unclear whether signaling pathways triggered by hypoxia contribute to radiation resistance. For example, intratumoral hypoxia can increase hypoxia inducible factor 1 alpha (HIF-1α), which may regulate pathways that contribute to radiation sensitization or radiation resistance. To clarify the role of HIF-1α in regulating tumor response to radiation, we generated a novel genetically engineered mouse model of soft tissue sarcoma with an intact or deleted HIF-1α. Deletion of HIF-1α sensitized primary sarcomas to radiation exposure in vivo. Moreover, cell lines derived from primary sarcomas lacking HIF-1α, or in which HIF-1α was knocked down, had decreased clonogenic survival in vitro, demonstrating that HIF-1α can promote radiation resistance in a cell autonomous manner. In HIF-1α-intact and -deleted sarcoma cells, radiation-induced reactive oxygen species, DNA damage repair and activation of autophagy were similar. However, sarcoma cells lacking HIF-1α had impaired mitochondrial biogenesis and metabolic response after irradiation, which might contribute to radiation resistance. These results show that HIF-1α promotes radiation resistance in a cell autonomous manner.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Sarcoma/metabolism , Sarcoma/radiotherapy , Animals , Cell Line, Tumor , Chemoradiotherapy , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mitochondria/metabolism , Mitochondria/radiation effects , Mitochondrial Size/genetics , Mitochondrial Size/radiation effects , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Sarcoma/genetics , Sarcoma/pathology , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/radiation effects
5.
Tsitol Genet ; 48(3): 43-7, 2014.
Article in Ukrainian | MEDLINE | ID: mdl-25016828

ABSTRACT

The ultrastructural organization of loach embryo cells (Misgurnus fossilis L) at the stage of the first and the tenth embryo divisions was investigated in the control and under the influence of low intensity helium-neon laser irradiation of 5 min exposure. The effect of laser irradiation led to ultrastructural changes in cell organelles, increasing the number and size of mitochondria, and as a result their shape changes. Under the influence of laser irradiation, the activation of cellular digestion processes took place, the number of vacuoles and lysosomes increased. The results explain the possible mechanism laser irradiation impact at the cellular level.


Subject(s)
Cypriniformes/embryology , Embryo, Nonmammalian , Lasers, Gas , Animals , Cell Membrane/radiation effects , Cell Membrane/ultrastructure , Dose-Response Relationship, Radiation , Embryo, Nonmammalian/radiation effects , Embryo, Nonmammalian/ultrastructure , Mitochondria/radiation effects , Mitochondria/ultrastructure , Mitochondrial Size/radiation effects
6.
PLoS One ; 9(6): e100738, 2014.
Article in English | MEDLINE | ID: mdl-24968221

ABSTRACT

Neoadjuvant chemoradiation therapy (CRT) is increasingly the standard of care for locally advanced oesophageal cancer. A complete pathological response to CRT is associated with a favourable outcome. Radiation therapy is important for local tumour control, however, radioresistance remains a substantial clinical problem. We hypothesise that alterations in mitochondrial function and energy metabolism are involved in the radioresistance of oesophageal adenocarcinoma (OAC). To investigate this, we used an established isogenic cell line model of radioresistant OAC. Radioresistant cells (OE33 R) demonstrated significantly increased levels of random mitochondrial mutations, which were coupled with alterations in mitochondrial function, size, morphology and gene expression, supporting a role for mitochondrial dysfunction in the radioresistance of this model. OE33 R cells also demonstrated altered bioenergetics, demonstrating significantly increased intracellular ATP levels, which was attributed to enhanced mitochondrial respiration. Radioresistant cells also demonstrated metabolic plasticity, efficiently switching between the glycolysis and oxidative phosphorylation energy metabolism pathways, which were accompanied by enhanced clonogenic survival. This data was supported in vivo, in pre-treatment OAC tumour tissue. Tumour ATP5B expression, a marker of oxidative phosphorylation, was significantly increased in patients who subsequently had a poor pathological response to neoadjuvant CRT. This suggests for the first time, a role for specific mitochondrial alterations and metabolic remodelling in the radioresistance of OAC.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Energy Metabolism/radiation effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Mitochondria/metabolism , Mitochondria/radiation effects , Radiation Tolerance , Adenocarcinoma/therapy , Adult , Aged , Cell Line, Tumor , Chemoradiotherapy, Adjuvant , Energy Metabolism/drug effects , Esophageal Neoplasms/therapy , Female , Humans , Male , Middle Aged , Mitochondria/drug effects , Mitochondrial Size/drug effects , Mitochondrial Size/radiation effects , Mutagenesis/drug effects , Mutagenesis/radiation effects , Radiation Tolerance/drug effects , Treatment Outcome
7.
Radiat Res ; 180(4): 360-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24033192

ABSTRACT

Free radicals generated by mitochondria are candidates for mediating long-lasting effects of radiation on cells, including genetic instability. To better understand the significance of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in these long-term effects we assayed ROS and RNS levels, the mitochondrial membrane potential and mass, and the frequency of DNA strand breaks, apoptosis and necrosis in human leukemic cells (K562 and HL60) after 12 Gy of X irradiation. An increase in intracellular ROS level was observed immediately post-irradiation, and about 24 h later a second increase of ROS was accompanied by increase in nitrogen oxide, mitochondrial potential and mitochondrial mass in both cell types. The second peak of ROS level was partially inhibited by rotenone, an inhibitor of mitochondrial complex I, in K562 but not in HL60 cells suggesting that the sources of ROS differed in the two cell types. The frequency of DNA breaks showed kinetics similar to ROS levels, with a sharp peak immediately after irradiation and a second increase 24 and 48 h later, which was significantly higher in K562 cells. Forty-eight hours after irradiation an increase in the frequency of apoptotic cells was observed in both cell lines, which became larger and statistically significant in K562 cells after inhibition of mitochondrial complex I. Our results show that ionizing radiation activates cellular processes which produce long-lasting ROS and RNS radicals, which may have different sources in different cell types and could participate in cellular signaling networks important for radiosensitivity and mode of cell death.


Subject(s)
Mitochondria/metabolism , Mitochondria/radiation effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , DNA Breaks/drug effects , DNA Breaks/radiation effects , Dose-Response Relationship, Radiation , Electron Transport Complex I/metabolism , HL-60 Cells , Humans , K562 Cells , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondrial Size/drug effects , Mitochondrial Size/radiation effects , Nitric Oxide/metabolism , Rotenone/pharmacology , Superoxides/metabolism , Time Factors
8.
Radiat Res ; 168(1): 134-42, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17722997

ABSTRACT

The bystander effect describes radiation-like damage in unirradiated cells either in the vicinity of irradiated cells or exposed to medium from irradiated cells. This study aimed to further characterize the poorly understood mitochondrial response to both direct irradiation and bystander factor(s) in human keratinocytes (HPV-G) and Chinese hamster ovarian cells (CHO-K1). Oxygen consumption rates were determined during periods of state 4, state 3 and uncoupled respiration. Mitochondrial mass was determined using MitoTracker FM. CHO-K1 cells showed significantly reduced oxygen consumption rates 4 h after exposure to 5 Gy direct radiation and irradiated cell conditioned medium (ICCM) and an apparent recovery 12-24 h later. The apparent recovery was likely due to the substantial increase in mitochondrial mass observed in these cells as soon as 4 h after exposure. HPV-G cells, on the other hand, showed a sustained increase in oxygen consumption rates after ICCM exposure and a transient increase 4 h after exposure to 5 Gy direct radiation. A significant increase in mitochondrial mass per HPV-G cell was observed after exposure to both direct radiation and ICCM. These findings are indicative of a stress response to mitochondrial dysfunction that increases the number of mitochondria per cell.


Subject(s)
Bystander Effect/radiation effects , Gamma Rays , Mitochondria/pathology , Mitochondria/radiation effects , Mitochondrial Size/radiation effects , Animals , Cell Line , Cricetinae , Humans , Oxygen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...