Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 8(9)2019 09 06.
Article in English | MEDLINE | ID: mdl-31489941

ABSTRACT

Liver damage upon exposure to ionizing radiation, whether accidental or because of therapy can contribute to liver dysfunction. Currently, radiation therapy is used for various cancers including hepatocellular carcinoma; however, the treatment dose is limited by poor liver tolerance to radiation. Furthermore, reliable biomarkers to predict liver damage and associated side-effects are unavailable. Here, we investigated fibrinogen-like 1 (FGL1)-expression in the liver and plasma after radiation exposure. We found that 30 Gy of liver irradiation (IR) induced cell death including apoptosis, necrosis, and autophagy, with fibrotic changes in the liver occurring during the acute and subacute phase in mice. Moreover, FGL1 expression pattern in the liver following IR was associated with liver damage represented by injury-related proteins and oxidative stress markers. We confirmed the association between FGL1 expression and hepatocellular injury by exposing human hepatocytes to radiation. To determine its suitability, as a potential biomarker for radiation-induced liver injury, we measured FGL1 in the liver tissue and the plasma of mice following total body irradiation (TBI) or liver IR. In TBI, FGL1 showed the highest elevation in the liver compared to other major internal organs including the heart, lung, kidney, and intestine. Notably, plasma FGL1 showed good correlation with radiation dose by liver IR. Our data revealed that FGL1 upregulation indicates hepatocellular injury in response to IR. These results suggest that plasma FGL1 may represent a potential biomarker for acute and subacute radiation exposure to the liver.


Subject(s)
Fibrinogen/metabolism , Liver Cirrhosis/blood , Liver/radiation effects , Radiation Injuries, Experimental/blood , Animals , Apoptosis , Autophagy , Biomarkers/blood , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/radiation effects , Humans , Liver/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Radiation Injuries, Experimental/pathology , Radiation, Ionizing
2.
Int J Mol Sci ; 19(8)2018 Aug 13.
Article in English | MEDLINE | ID: mdl-30104472

ABSTRACT

Although many attempts have been made to improve the efficacy of radiotherapy to treat cancer, radiation resistance is still an obstacle in lung cancer treatment. Oridonin is a natural compound with promising antitumor efficacy that can trigger cancer cell death; however, its direct cellular targets, efficacy as a radiosensitizer, and underlying mechanisms of activity remain unclear. Herein, we report that oridonin exhibits additive cytotoxic and antitumor activity with radiation using the H460 non-small cell lung cancer cell lines. We assessed the effect of oridonin by proliferation, clonogenic, reactive oxygen species (ROS) production, DNA damage, and apoptosis assays. In vitro, oridonin enhanced the radiation-induced inhibition of cell growth and clonogenic survival. Oridonin also facilitated radiation-induced ROS production and DNA damage and enhanced apoptotic cell death. In vivo, the combination of oridonin and radiation effectively inhibited H460 xenograft tumor growth, with higher caspase-3 activation and H2A histone family member X (H2AX) phosphorylation compared with that of radiation alone. Our findings suggest that oridonin possesses a novel mechanism to enhance radiation therapeutic responses by increasing DNA damage and apoptosis. In conclusion, oridonin may be a novel small molecule to improve radiotherapy in non-small cell lung cancer.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , Diterpenes, Kaurane/pharmacology , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Caspase 3/metabolism , Cell Line, Tumor , DNA Damage/radiation effects , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/therapeutic use , Female , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Transplantation, Heterologous
3.
Int J Mol Sci ; 19(7)2018 Jul 19.
Article in English | MEDLINE | ID: mdl-30029554

ABSTRACT

The expansion of mobile phone use has raised questions regarding the possible biological effects of radiofrequency electromagnetic field (RF-EMF) exposure on oxidative stress and brain inflammation. Despite accumulative exposure of humans to radiofrequency electromagnetic fields (RF-EMFs) from mobile phones, their long-term effects on oxidative stress and neuroinflammation in the aging brain have not been studied. In the present study, middle-aged C57BL/6 mice (aged 14 months) were exposed to 1950 MHz electromagnetic fields for 8 months (specific absorption rate (SAR) 5 W/kg, 2 h/day, 5 d/week). Compared with those in the young group, levels of protein (3-nitro-tyrosine) and lipid (4-hydroxy-2-nonenal) oxidative damage markers were significantly increased in the brains of aged mice. In addition, levels of markers for DNA damage (8-hydroxy-2'-deoxyguanosine, p53, p21, γH2AX, and Bax), apoptosis (cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase 1 (PARP-1)), astrocyte (GFAP), and microglia (Iba-1) were significantly elevated in the brains of aged mice. However, long-term RF-EMF exposure did not change the levels of oxidative stress, DNA damage, apoptosis, astrocyte, or microglia markers in the aged mouse brains. Moreover, long-term RF-EMF exposure did not alter locomotor activity in aged mice. Therefore, these findings indicate that long-term exposure to RF-EMF did not influence age-induced oxidative stress or neuroinflammation in C57BL/6 mice.


Subject(s)
Aging/pathology , Brain/pathology , Electromagnetic Fields , Inflammation/pathology , Oxidative Stress/radiation effects , Radio Waves , Animals , Behavior, Animal , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , DNA Damage , Glial Fibrillary Acidic Protein/metabolism , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Motor Activity , Poly(ADP-ribose) Polymerases/metabolism
4.
Int J Mol Sci ; 18(10)2017 Oct 07.
Article in English | MEDLINE | ID: mdl-28991157

ABSTRACT

Radiation-induced intestinal toxicity is common among cancer patients after radiotherapy. Endothelial cell dysfunction is believed to be a critical contributor to radiation tissue injury in the intestine. Geranylgeranylacetone (GGA) has been used to treat peptic ulcers and gastritis. However, the protective capacity of GGA against radiation-induced intestinal injury has not been addressed. Therefore, we investigated whether GGA affects intestinal damage in mice and vascular endothelial cell damage in vitro. GGA treatment significantly ameliorated intestinal injury, as evident by intestinal crypt survival, villi length and the subsequently prolonged survival time of irradiated mice. In addition, intestinal microvessels were also significantly preserved in GGA-treated mice. To clarify the effect of GGA on endothelial cell survival, we examined endothelial function by evaluating cell proliferation, tube formation, wound healing, invasion and migration in the presence or absence of GGA after irradiation. Our findings showed that GGA plays a role in maintaining vascular cell function; however, it does not protect against radiation-induced vascular cell death. GGA promoted endothelial function during radiation injury by preventing the loss of VEGF/VEGFR1/eNOS signaling and by down-regulating TNFα expression in endothelial cells. This finding indicates the potential impact of GGA as a therapeutic agent in mitigating radiation-induced intestinal damage.


Subject(s)
Diterpenes/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Animals , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Wound Healing/drug effects
5.
Oncotarget ; 8(41): 69386-69397, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29050211

ABSTRACT

Radiotherapy is a common treatment for cancer patients, but its use is often restricted by the tolerance of normal tissue. As cancer patients live longer, delayed radiation effects on normal tissue have become a concern. Radiation-induced enteropathy, including inflammatory bowel disease and fibrosis, are major issues for long-term cancer survivors. To investigate whether silibinin attenuates delayed radiation-induced intestinal injury in mice, we focused on intestinal fibrotic changes. Silibinin improved delayed radiation injuries in mice in association with decreased collagen deposition within the intestines and deceased transforming growth factor (TGF)-ß1 levels in the intestine and plasma. Treating mice bearing CT26 mouse colon cancer tumors with both silibinin and radiation stimulated tumor regression more than radiation alone. We also investigated the effect of silibinin on the radiation-induced epithelial-to-mesenchymal transition (EMT), the primary mechanism of fibrosis. We assessed changes in E-cadherin, N-cadherin, and α-smooth muscle actin expression, and demonstrated that silibinin attenuates radiation-induced EMT. Irradiating intestinal epithelial cells increased TGF-ß1 levels, but silibinin suppressed TGF-ß1 expression by inhibiting Smad2/3 phosphorylation. These results suggest silibinin has the potential to serve as a useful therapeutic agent in patients with radiation-induced intestinal fibrosis.

6.
Aging Cell ; 16(4): 773-784, 2017 08.
Article in English | MEDLINE | ID: mdl-28514055

ABSTRACT

Paradoxical observations have been made regarding the role of caveolin-1 (Cav-1) during cellular senescence. For example, caveolin-1 deficiency prevents reactive oxygen species-induced cellular senescence despite mitochondrial dysfunction, which leads to senescence. To resolve this paradox, we re-addressed the role of caveolin-1 in cellular senescence in human diploid fibroblasts, A549, HCT116, and Cav-1-/- mouse embryonic fibroblasts. Cav-1 deficiency (knockout or knockdown) induced cellular senescence via a p53-p21-dependent pathway, downregulating the expression level of the cardiolipin biosynthesis enzymes and then reducing the content of cardiolipin, a critical lipid for mitochondrial respiration. Our results showed that Cav-1 deficiency decreased mitochondrial respiration, reduced the activity of oxidative phosphorylation complex I (CI), inactivated SIRT1, and decreased the NAD+ /NADH ratio. From these results, we concluded that Cav-1 deficiency induces premature senescence via mitochondrial dysfunction and silent information regulator 2 homologue 1 (SIRT1) inactivation.


Subject(s)
Caveolin 1/genetics , Cellular Senescence/genetics , Fibroblasts/metabolism , Mitochondria/metabolism , Sirtuin 1/genetics , A549 Cells , Animals , Cardiolipins/biosynthesis , Caveolin 1/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian , Fibroblasts/pathology , Gene Expression Regulation , HCT116 Cells , Humans , Mice , Mitochondria/pathology , NAD/metabolism , Oxidative Phosphorylation , Primary Cell Culture , Signal Transduction , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Int J Nanomedicine ; 11: 1413-25, 2016.
Article in English | MEDLINE | ID: mdl-27103799

ABSTRACT

Investigation of potential therapeutics for targeting breast cancer stem cells (BCSCs) is important because these cells are regarded as culprit of breast cancer relapse. Accomplishing this kind of strategy requires a specific drug-delivery system using the distinct features of liposomes. Studies on targeted liposomal delivery systems have indicated the conjugation of hyaluronan (HA), a primary ligand for CD44 surface markers, as an appropriate method for targeting BCSCs. For this study, enriched BCSCs were obtained by culturing MCF-7 breast cancer cells in nonadherent conditions. The enriched BCSCs were challenged with HA-conjugated liposomes encapsulating gemcitabine (2, 2-difluoro-2-deoxycytidine, GEM). In vitro study showed that the HA-conjugated liposomes significantly enhanced the cytotoxicity, anti-migration, and anti-colony formation abilities of GEM through targeting of CD44 expressed on BCSCs. In pharmacokinetic study, area under the drug concentration vs time curve (AUC) of the immunoliposomal GEM was 3.5 times higher than that of free GEM, indicating that the HA-conjugated liposomes enhanced the stability of GEM in the bloodstream and therefore prolonged its half-life time. The antitumor effect of the immunoliposomal GEM was 3.3 times higher than that of free GEM in a xenograft mouse model, probably reflecting the unique targeting of the CD44 receptor by HA and the increased cytotoxicity and stability through the liposomal formulation. Furthermore, marginal change in body weight demonstrated that the use of liposomes considerably reduced the systemic toxicity of GEM on normal healthy cells. Taken together, this study demonstrates that HA-conjugated liposomes encapsulating GEM show promise for the therapy of breast cancer in vitro and in a xenograft model by targeting the BCSCs.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Hyaluronic Acid/chemistry , Liposomes/chemistry , Neoplastic Stem Cells/drug effects , Animals , Antimetabolites, Antineoplastic/chemistry , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Delivery Systems , Female , Half-Life , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays , Gemcitabine
8.
Oncol Rep ; 35(2): 841-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26717900

ABSTRACT

Cellular senescence is a state of irreversible growth arrest that can be triggered by multiple mechanisms, including telomere shortening, the epigenetic derepression of the INK4α/ARF locus and DNA damage. Senescence has been considered a tumor­suppressing mechanism that permanently arrests cells at risk for malignant transformation. However, accumulating evidence shows that senescent cells have deleterious effects on the tissue microenvironment. Some of these effects could be attributed to the senescence­associated secretory phenotype that has the ability to promote tumor progression. However, secreted proteins from senescent tumor cells and their effects on the tumor microenvironment due to ionizing radiation (IR) exposure have not yet been fully elucidated. In the present study, we analyzed cytokines secreted from IR­induced senescent MCF7 cells by using cytokine microarrays and confirmed by western blot analysis that increased secretion of osteoprotegerin (OPG), midkine (MDK) and apolipoprotein E3 (ApoE3) occurs in these cells. Invasive, migratory and wound­healing activities were observed in MDA­MB­231 and MCF­10A cells following treatment with recombinant human OPG, MDK and ApoE3 proteins. Additionally, tube­formation activity was assessed in OPG­, MDK­ and ApoE3­treated human umbilical vein endothelial cells (HUVECs). We found that OPG, MDK and ApoE3 affected cell motility and tube­formation activity. Since OPG markedly affected cell motility, we examined the effect of senescent conditioned media containing neutralizing OPG antibodies on migration and wound­healing activity. Our results demonstrated that IR­induced senescent tumor cells influence the tumor microenvironment by increasing the production of cytokines, such as OPG, MDK and ApoE3. Furthermore, these data suggest that OPG is likely a promising target capable of reducing the deleterious effects on the tumor microenvironment during radiation therapy.


Subject(s)
Cellular Senescence/physiology , Cellular Senescence/radiation effects , Cytokines/biosynthesis , Tumor Microenvironment/physiology , Blotting, Western , Cell Line, Tumor , Humans , Oligonucleotide Array Sequence Analysis , Radiation, Ionizing
9.
Proteomics ; 12(18): 2822-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833545

ABSTRACT

Cellular senescence is a physiological program of irreversible growth arrest that is considered to play an important role in tumor suppression. Recent studies demonstrated that senescent cells secrete multiple growth regulatory proteins that could alter the behavior of neighboring cells. In this study, we investigated the effect of secretory proteins from ionizing radiation (IR) induced senescent tumor cells on normal and tumor cells. Conditioned medium (CM) from IR-induced senescent MCF7 cells significantly increased cell proliferation, invasion, migration, and wound healing activity in MCF7 cells and HUVECs. Comparative proteomics analysis revealed 24 differentially secreted protein spots including Raf kinase inhibitor protein (RKIP), α-Enolase, AKAP9, and MARK4, and the findings were confirmed by Western blot analysis of IR-induced senescent cancer cells. We found that RKIP was secreted via the classical pathway, and the transfection of small interfering RNA against RKIP suppressed CM-induced migration in MCF7 cells. Treatment with recombinant human RKIP increased the migratory activity of MCF7 cells. Taken together, our results demonstrate that the senescence-associated secretory protein RKIP could be the principal target to prevent the potential effects of the secretome from IR-induced senescent tumor cells on neighboring cell migration.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement/radiation effects , Cellular Senescence/radiation effects , Phosphatidylethanolamine Binding Protein/metabolism , Proteome/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/radiation effects , Female , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Phosphatidylethanolamine Binding Protein/genetics , RNA, Small Interfering , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection
10.
J Radiat Res ; 53(2): 176-83, 2012.
Article in English | MEDLINE | ID: mdl-22510589

ABSTRACT

Exposure to environmental stressors can be measured by monitoring the cellular stress response in target cells. Here, we used the cellular stress response to investigate whether single or combined radio frequency (RF) radiation could induce stress response in human cells. Cellular stress responses in MCF10A human breast epithelial cells were characterized after exposure to 4 h of RF radiation [code division multiple access (CDMA) or CDMA plus wideband CDMA (WCDMA)] or 2 h RF radiation on 3 consecutive days. Specific absorption rate (SAR) was 4.0 W/kg for CDMA signal alone exposure and 2.0 W/kg each, 4.0 W/kg in total for combined CDMA plus WCDMA signals. Expression levels and phosphorylation states of specific heat shock proteins (HSPs) and mitogen-activated protein kinases (MAPKs) were analyzed by Western blot. It was found that HSP27 and ERK1/2 phosphorylations are the most sensitive markers of the stress response in MCF10A cells exposed to heat shock or ionizing radiation. Using these markers, we demonstrated that neither one-time nor repeated single (CDMA alone) or combined (CDMA plus WCDMA) RF radiation exposure significantly altered HSP27 and ERK1/2 phosphorylations in MCF10A cells (p > 0.05). The lack of a statistically significant alteration in HSP27 and ERK1/2 phosphorylations suggests that single or combined RF radiation exposure did not elicit activation of HSP27 and ERK1/2 in MCF10A cells.


Subject(s)
Breast/physiology , Breast/radiation effects , Heat-Shock Proteins/metabolism , Microwaves , Radio Waves , Stress, Physiological/physiology , Stress, Physiological/radiation effects , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Radiation Dosage
11.
J Radiat Res ; 53(1): 79-86, 2012.
Article in English | MEDLINE | ID: mdl-22302048

ABSTRACT

The aim of this study was to determine whether extremely low frequency magnetic fields (ELF-MF) could affect intracellular reactive oxygen species (ROS) levels and antioxidant enzyme activity. After MCF10A human breast epithelial cells were exposed to 1 mT of 60 Hz ELF-MF for 4 hours, intracellular ROS level, superoxide dismutase (SOD) activity, and reduced to oxidized glutathione (GSH/GSSG) ratio were measured. The cells exposed to ELF-MF did not evidence statistically significant changes in the above-mentioned biological parameters as compared to either the incubator controls or sham-exposed cells. By way of contrast, the IR-exposed cells exhibited marked changes in ROS level, SOD activity, and GSH/GSSG ratio. When we assessed morphological changes and senescence-associated beta-galactosidase (SA-ß-Gal) activity, only the IR-exposed cells were positive. According to our results, it could be concluded that ELF-MF has no effect on intracellular ROS level, SOD activity, and GSH/GSSG ratio under our exposure condition.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Magnetic Fields/adverse effects , Oxidative Stress , Cell Line, Tumor/radiation effects , Cell Shape , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Equipment Design , Female , Gamma Rays/adverse effects , Glutathione/metabolism , Humans , Neoplasm Proteins/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Research Design , Superoxide Dismutase/metabolism
12.
Bioelectromagnetics ; 32(3): 169-78, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21365661

ABSTRACT

The aim of this study was to investigate whether single or combined radio frequency (RF) radiation exposure has effects on the cell cycle and its regulatory proteins. Exposure of MCF7 cells to either single (837 MHz) or combined (837 and 1950 MHz) RF radiation was conducted at specific absorption rate values of 4 W/kg for 1 h. During the exposure period, the chamber was made isothermal by circulating water through the cavity. After RF radiation exposure, DNA synthesis rate and cell cycle distribution were assessed. The levels of cell cycle regulatory proteins, p53, p21, cyclins, and cyclin-dependent kinases were also examined. The positive control group was exposed to 0.5 and 4 Gy doses of ionizing radiation (IR) and showed changes in DNA synthesis and cell cycle distribution. The levels of p53, p21, cyclin A, cyclin B1, and cyclin D1 were also affected by IR exposure. In contrast to the IR-exposed group, neither the single RF radiation- nor the combined RF radiation-exposed group elicited alterations in DNA synthesis, cell cycle distribution, and levels of cell cycle regulatory proteins. These results indicate that neither single nor combined RF radiation affect cell cycle progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/radiation effects , Radio Waves , Bromodeoxyuridine/metabolism , Cell Line, Tumor , DNA/biosynthesis , Humans
13.
Oncol Rep ; 24(2): 395-403, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20596626

ABSTRACT

Senescence has been suggested as a defense mechanism to block sporadic induction of cancer cells. Radiation treatment induces proliferating cancer cells to turn into non-proliferating senescent cells in vitro. To characterize transcriptional reprogramming after radiation treatment, we measured the gene expression profiles of MCF7 at different time points after treatment. In these experiments, we found that IR induced premature senescence in MCF7 cells, and IR treatment resulted in significant changes in the expression of 305 marker genes (<1% FDR), which were strongly correlated (|r|>0.9) with IR treatment in a time-dependent manner. Functional analysis of these markers indicated that the dynamics of cytoskeletal structure and lysosomal activity gradually increased. The expression of maker genes for modulator proteins, that were responsible for the dynamics of actin stress fibers and focal adhesion, displayed a particularly strong positive correlation with senescence-associated (SA) morphological changes through time. We also observed a strong induction of genes related to lysosomal metabolic activity, which was accompanied by an increase in the number of SA-beta-Gal positive cells. However, the expression of genes for cell cycle progression, post-transcription and translation activities gradually decreased after radiation treatment. Especially, we observed clear cell cycle arrest specifically at the S and G2/M phases with consistent down-regulation of genes for microtubule assembly/disassembly or spindle biogenesis.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Carcinoma/genetics , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Actins/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Shape/genetics , Cell Shape/radiation effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lysosomes/metabolism , Lysosomes/radiation effects , Protein Multimerization/genetics , Protein Multimerization/radiation effects , Radiation, Ionizing , Time Factors
14.
J Radiat Res ; 51(2): 205-13, 2010.
Article in English | MEDLINE | ID: mdl-20339255

ABSTRACT

Although many in vitro studies have previously been conducted to elucidate the biological effects of radio frequency (RF) radiation over the past decades, the existence and nature of any effects is still inconclusive. In an effort to further elucidate this question, we have monitored changes in protein expression profiles in RF-exposed MCF7 human breast cancer cells using two-dimensional gel electrophoresis. MCF7 cells were exposed to 849 MHz RF radiation for 1 h per day for three consecutive days at specific absorption rates (SARs) of either 2 W/Kg or 10 W/kg. During exposure, the temperature in the exposure chamber was kept in an isothermal condition. Twenty-four hours after the final RF exposure, the protein lysates from MCF cells were prepared and two-dimensional electrophoretic analyses were conducted. The protein expression profiles of the MCF cells were not significantly altered as the result of RF exposure. None of the protein spots on the two-dimensional electrophoretic gels showed reproducible changes in three independent experiments. To determine effect of RF radiation on protein expression profiles more clearly, three spots showing altered expression without reproducibility were identified using electrospray ionization tandem mass spectrometry analysis and their expressions were examined with RT-PCR and Western blot assays. There was no alteration in their mRNA and protein levels. As we were unable to observe any significant and reproducible changes in the protein expression profiles of the RF radiation-exposed MCF7 cells using high throughput and non-high throughput techniques, it seems unlikely that RF exposure modulates the protein expression profile.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation, Neoplastic/radiation effects , Neoplasm Proteins/analysis , Radio Waves , Adenocarcinoma/chemistry , Blotting, Western , Breast Neoplasms/chemistry , Cell Line, Tumor/chemistry , Cell Line, Tumor/radiation effects , Cell Phone , Female , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
15.
Cancer Res ; 69(11): 4638-47, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487283

ABSTRACT

Induction of premature senescence may be a promising strategy for cancer treatment. However, biomarkers for senescent cancer cells are lacking. To identify such biomarkers, we performed comparative proteomic analysis of MCF7 human breast cancer cells undergoing cellular senescence in response to ionizing radiation (IR). IR-induced senescence was associated with up-regulation of cathepsin D (CD) and down-regulation of eukaryotic translation elongation factor 1beta2 (eEF1B2), as confirmed by Western blot. The other elongation factor, eukaryotic translation elongation factor 1alpha1 (eEF1A1), was also down-regulated. IR-induced senescence was associated with similar changes of CD and eEF1 (eEF1A1 and eEF1B2) levels in the HCT116 colon cancer cell line and the H460 lung cancer cell line. Up-regulation of CD and down-regulation of eEF1 seemed to be specific to senescence, as they were observed during cellular senescence induced by hydrogen peroxide or anticancer drugs (camptothecin, etoposide, or 50 ng doxorubicin) but not during apoptosis induced by Taxol or 10 microg doxorubicin or autophagy induced by tamoxifen. The same alterations in CD and eEF1A1 levels were observed during replicative senescence and Ras oncogene-induced senescence. Transient cell cycle arrest did not alter levels of eEF1 or CD. Chemical inhibition of CD (pepstatin A) and small interfering RNA-mediated knockdown of CD and eEF1 revealed that these factors participate in cell proliferation. Finally, the senescence-associated alteration in CD and eEF1 levels observed in cell lines was also observed in IR-exposed xenografted tumors. These findings show that CD and eEF1 are promising markers for the detection of cellular senescence induced by a variety of treatments.


Subject(s)
Cathepsin D/physiology , Cellular Senescence , Peptide Elongation Factor 1/physiology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers/metabolism , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Gamma Rays/adverse effects , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Elongation Factor 1/antagonists & inhibitors , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , RNA, Small Interfering/pharmacology , Radiation Dosage , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...