Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Proc Natl Acad Sci U S A ; 119(43): e2208506119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36256824

ABSTRACT

DNA-damaging treatments such as radiotherapy (RT) have become promising to improve the efficacy of immune checkpoint inhibitors by enhancing tumor immunogenicity. However, accompanying treatment-related detrimental events in normal tissues have posed a major obstacle to radioimmunotherapy and present new challenges to the dose delivery mode of clinical RT. In the present study, ultrahigh dose rate FLASH X-ray irradiation was applied to counteract the intestinal toxicity in the radioimmunotherapy. In the context of programmed cell death ligand-1 (PD-L1) blockade, FLASH X-ray minimized mouse enteritis by alleviating CD8+ T cell-mediated deleterious immune response compared with conventional dose rate (CONV) irradiation. Mechanistically, FLASH irradiation was less efficient than CONV X-ray in eliciting cytoplasmic double-stranded DNA (dsDNA) and in activating cyclic GMP-AMP synthase (cGAS) in the intestinal crypts, resulting in the suppression of the cascade feedback consisting of CD8+ T cell chemotaxis and gasdermin E-mediated intestinal pyroptosis in the case of PD-L1 blocking. Meanwhile, FLASH X-ray was as competent as CONV RT in boosting the antitumor immune response initiated by cGAS activation and achieved equal tumor control in metastasis burdens when combined with anti-PD-L1 administration. Together, the present study revealed an encouraging protective effect of FLASH X-ray upon the normal tissue without compromising the systemic antitumor response when combined with immunological checkpoint inhibitors, providing the rationale for testing this combination as a clinical application in radioimmunotherapy.


Subject(s)
Neoplasms , Radioimmunotherapy , Mice , Animals , X-Rays , Pyroptosis , Immune Checkpoint Inhibitors , Ligands , Nucleotidyltransferases/metabolism
3.
Free Radic Biol Med ; 162: 514-522, 2021 01.
Article in English | MEDLINE | ID: mdl-33197538

ABSTRACT

Two-deoxy-d-glucose (2-DG) mediated glucose restriction (GR) has been applied as a potential therapeutic strategy for tumor clinical treatments. However, increasing evidences have indicated that 2-DG alone is inefficient in killing tumor cells, and the effect of 2-DG on modifying tumor radio-responses also remains controversial. In this study, we found that 2-DG triggered metabolic adaption in U87 glioma cells by up-regulating nicotinamide phosphoribosyltransferase (NAMPT) and cellular NAD+ content, which abolished 2-DG-induced potential radiosensitizing effect in glioma cells. Strikingly, NAD+ depletion evoked notable oxidative stress by NADPH reduction and hence re-radiosensitized 2-DG-treated glioma cells. Furthermore, isocitrate dehydrogenase-1 (IDH1) mutant U87 glioma cells with deficiency in the rate-limiting enzyme of Preiss-Handler pathway nicotinate phosphoribosyltransferase (Naprt1) revealed notable 2-DG-induced oxidative stress and radiosensitization. Our findings implied that targeting NAD+ could radiosensitize gliomas with GR, and 2-DG administration could be benefit for tumor patients with IDH1 mutation.


Subject(s)
Glioma , NAD , Cell Line, Tumor , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Isocitrate Dehydrogenase/genetics , Mutation , NADP
4.
Cell Death Dis ; 11(7): 501, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632140

ABSTRACT

During radiologic or nuclear accidents, high-dose ionizing radiation (IR) can cause gastrointestinal syndrome (GIS), a deadly disorder that urgently needs effective therapy. Unfortunately, current treatments based on natural products and antioxidants have shown very limited effects in alleviating deadly GIS. Reserve intestinal stem cells (ISCs) and secretory progenitor cells are both reported to replenish damaged cells and contribute to crypt regeneration. However, the suppressed ß-catenin/c-MYC axis within these slow-cycling cells leads to limited regenerative response to restore intestinal integrity during fatal accidental injury. Current study demonstrates that post-IR overexpression of TIGAR, a critical downstream target of c-MYC in mouse intestine, mounts a hyperplastic response in Bmi1-creERT+ reserve ISCs, and thus rescues mice from lethal IR exposure. Critically, by eliminating damaging reactive oxygen species (ROS) yet retaining the proliferative ROS signals, TIGAR-overexpression enhances the activity of activator protein 1, which is indispensable for initiating reserve-ISC division after lethal radiation. In addition, it is identified that TIGAR-induction exclusively gears the Lgr5- subpopulation of reserve ISCs to regenerate crypts, and intestinal TIGAR-overexpression displays equivalent intestinal reconstruction to reserve-ISC-restricted TIGAR-induction. Our findings imply that precise administrations toward Lgr5- reserve ISCs are promising strategies for unpredictable lethal injury, and TIGAR can be employed as a therapeutic target for unexpected radiation-induced GIS.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Intestines/cytology , Phosphoric Monoester Hydrolases/metabolism , Radiation, Ionizing , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Stem Cells/metabolism , Stem Cells/radiation effects , Transcription Factor AP-1/metabolism , Animals , Female , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/pathology , Male , Mice , Models, Biological , Reactive Oxygen Species/metabolism , Regeneration/radiation effects
5.
Oncol Lett ; 19(2): 1322-1330, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31966064

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is the most frequently mutated gene in World Health Organization grade II-III and secondary glioma. The majority of IDH1 mutation cases involve the substitution from arginine to histidine at codon 132 (IDH1-R132H). Although the oncogenic role of IDH1-R132H has been confirmed, patients with IDH1-R132H brain tumors exhibit a better response to radiotherapy compared with those with wild-type (WT) IDH1. In the present study, the potential mechanism of radiosensitization mediated by IDH1-R132H was investigated by overexpressing IDH1-R132H in U87MG glioma cells. The results demonstrated decreased clonogenic capacity of IDH1-R132H-expressing cells, as well as delayed repair of DNA double-strand breaks compared with IDH1-WT. Data from The Cancer Genome Atlas were analyzed, which demonstrated that the expression of TP53-induced glycolysis and apoptosis regulator (TIGAR) was lower in patients with glioma harboring IDH1 mutations compared with that in patients with IDH1-WT. TIGAR-knockdown increases the radiosensitivity of glioma cells; in U87MG cells, IDH1-R132H suppressed TIGAR expression. Chromatin immunoprecipitation assays revealed increased levels of repressive H3K9me3 markers at the TIGAR promoter in IDH1-R132H compared with IDH1-WT. These data indicated that IDH1-R132H may overcome radioresistance in glioma cells through epigenetic suppression of TIGAR expression. However, these favorable effects were not observed in U87MG glioma stem-like cells. The results of the present study provide an improved understanding of the functionality of IDH1 mutations in glioma cells, which may improve the therapeutic efficacy of radiotherapy.

6.
Anticancer Res ; 39(11): 5919-5925, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31704816

ABSTRACT

BACKGROUND/AIM: The aim of the current study was to investigate the synergistic efficacy of Robo1 bichimeric antigen receptor-natural killer cell (BiCAR-NK) immunotherapy and 125I seed brachytherapy in an orthotopic pancreatic cancer mouse model. MATERIALS AND METHODS: The orthotopic pancreatic tumor model was established with human pancreatic cancer BxPC-3 cells expressing red fluorescent protein. The mice were treated with 125I seed implantation alone or the combination of 125I seeds with Robo1-specific CAR-NK cells. To assess tumor inhibition, in vivo fluorescence imaging was conducted. 7 Tesla magnetic resonance (7T-MR) scanning was applied to measure the changes in the metabolic profiles of tumor tissues. RESULTS: Tumor size was significantly reduced in the 125I and 125I +CAR-NK treated group compared to the untreated group (p<0.05). The 125I seed +CAR-NK treated group showed significantly higher tumor reduction than 125I seed treatment alone (p<0.05). T1 diffusion weighted imaging (T1DWI) sequence showed that the tumors of the 125I +BiCAR-NK treated group had a significantly higher grey scale value than the tumors from the untreated control and the group treated with 125I seed alone (p<0.05). CONCLUSION: Robo1 specific CAR-NK immunotherapy enhances efficacy of 125I seed brachytherapy in an orthotopic pancreatic cancer mouse model.


Subject(s)
Brachytherapy/methods , Immunotherapy , Iodine Radioisotopes/therapeutic use , Killer Cells, Natural/immunology , Nerve Tissue Proteins/immunology , Pancreatic Neoplasms/therapy , Receptors, Antigen/immunology , Receptors, Immunologic/immunology , Animals , Apoptosis , Cell Proliferation , Cytotoxicity, Immunologic/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Roundabout Proteins
7.
Technol Cancer Res Treat ; 18: 1533033819842546, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30961474

ABSTRACT

OBJECTIVES: Bortezomib has been widely used to treat multiple myeloma and other hematological malignancies. However, not much is known about its effect on solid tumors. The aim of this study was to study the effect of Bortezomib on human esophageal cancer cell lines and investigate the potential target pathways. METHODS: Two human esophageal cancer cell lines, TE-1 and KYSE-150, were used in this study. Cell viability, cell cycle distribution, and apoptosis after Bortezomib treatment was detected by Cell Counting Kit-8, flow cytometry, and Annexin V/propidium iodide staining, respectively. The genes targeted by Bortezomib were analyzed at the messenger RNA level by microarray chips and quantitative real-time polymerase chain reaction. RESULTS: The proliferation of human esophageal cancer cell lines was inhibited by Bortezomib in a dose- and time-dependent manner. Bortezomib treatment led to G2/M arrest and apoptosis. Microarray chips revealed multiple signaling pathways targeted by Bortezomib, including proteasome, endoplasmic reticulum, Wnt-, and calcium-mediated pathway. The expression patterns of 4 representative genes UBD, CUL3, HDAC6, and GADD45A were verified by quantitative real-time polymerase chain reaction and showed consistency with the microarray assay. CONCLUSION: Bortezomib could suppress cell viability, cause G2/M arrest, and induce apoptosis in human esophageal cancer cells, with possible targets including UBD, CUL3, HDAC6, and GADD45A.


Subject(s)
Bortezomib/pharmacology , Carcinoma/drug therapy , Cell Proliferation/drug effects , Esophageal Neoplasms/drug therapy , Apoptosis/drug effects , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cullin Proteins/genetics , Epithelial Cells/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 6/genetics , Humans , Microarray Analysis , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Ubiquitins/genetics , Wnt Signaling Pathway/drug effects
8.
DNA Repair (Amst) ; 70: 67-71, 2018 10.
Article in English | MEDLINE | ID: mdl-30212742

ABSTRACT

Homologous recombination (HR) and non-homologous end joining (NHEJ) are the two major mechanisms for the repair of DNA double-strand breaks (DSBs) in eukaryotic cells. Previously, we designed an assay for detecting NHEJ activity by using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system, however, this approach cannot be used to predict the activity of HR repair. Hence, we developed a novel method that is capable of quantitatively measuring both HR and NHEJ activities via CRISPR/Cas9-induced oligodeoxynucleotide (ODN)-mediated DSB repair. In the present experimental procedures, the CRISPR/Cas9 plasmid was cotransfected with single-stranded ODN (ssODN) or blunt-ended double-stranded ODN (dsODN), both of which harbored a unique marker sequence. After the induction of site-specific DSBs by CRISPR/Cas9 system, the ssODN, functioned as the donor template for HR repair, could insert the marker sequence into the DSB sites, while the dsODN was embedded in the DSB sites through NHEJ pathway. Next, by means of PCR analysis using a specific primer for the marker sequence and the primers that flank the DSB sites, the relative amount of integrated marker sequence in the genomic DNA could be quantitatively determined. The correlation between the marker sequence abundance and the HR and NHEJ activities was confirmed by using the selective HR and NHEJ inhibitors. This accessible and rapid quantitative assay for HR and NHEJ activities might be useful for the future research of the DSB repair mechanisms.


Subject(s)
CRISPR-Cas Systems/genetics , DNA End-Joining Repair/genetics , Homologous Recombination/genetics , Oligodeoxyribonucleotides/metabolism , Base Sequence , Cell Line , DNA Breaks, Double-Stranded , Humans
9.
Drug Des Devel Ther ; 12: 2223-2231, 2018.
Article in English | MEDLINE | ID: mdl-30038488

ABSTRACT

INTRODUCTION: In this study, the radiation-enhancing effects of combined treatment with nimotuzumab, a humanized EGFR-blocking antibody, and celecoxib, a COX-2 selective inhibitor, in human nasopharyngeal carcinoma (NPC) cells were investigated. MATERIALS AND METHODS: 3-(4,5-Dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide and clonogenic survival assays were done to evaluate the combined cytotoxic and radiosensitizing effects of nimotuzumab or celecoxib or the combination on CNE1 and CNE2 cells. Western blot analysis was performed to identify the effect of nimotuzumab and/or celecoxib with or without irradiation on the cytoplasmic and nuclear EGFR signaling pathways in CNE2 cells. RESULTS: Our results demonstrated that concurrent administration of nimotuzumab and celecoxib cooperatively enhanced the cytotoxicity and radiosensitivity of CNE2 cells but not CNE1 cells. The combination of both drugs with or without irradiation also cooperatively inhibited cytoplasmic and nuclear EGFR signaling pathways in CNE2 cells. CONCLUSION: Our results suggest a promising approach for the treatment of poorly differentiated NPC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Celecoxib/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Nasopharyngeal Carcinoma/drug therapy , Radiation-Sensitizing Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemical synthesis , Antineoplastic Combined Chemotherapy Protocols/chemistry , Celecoxib/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured
10.
Mol Cancer Ther ; 17(2): 419-431, 2018 02.
Article in English | MEDLINE | ID: mdl-28864683

ABSTRACT

Non-homologous end joining (NHEJ) is the major pathway responsible for the repair of ionizing radiation (IR)-induced DNA double-strand breaks (DSB), and correspondingly regulates the cellular response to IR. Identification of NHEJ inhibitors could substantially enhance the tumor radiosensitivity and improve the therapeutic efficiency of radiotherapy. In this study, we demonstrated a screening for NHEJ inhibitors using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system and high-resolution melting (HRM) analysis. Because NHEJ is regarded as an error-prone mechanism, the NHEJ-mediated ligation of the site-specific DSB induced by Cas9 nuclease would eventually cause the mutation of the targeted sequence. Then, HRM analysis, a reliable and rapid assay for detecting sequence variation, was performed to evaluate the mutation efficiency of the targeted site. Validating analysis confirmed the NHEJ activities were positively correlated with the mutation frequencies. Next, an approved drug library containing 1,540 compounds was interrogated by using this screening strategy. Our results identified ouabain, a cardiotonic agent, and penfluridol, an antipsychotic agent, have the capacity to restrain NHEJ activity. Further experiments in vitro revealed the radiosensitizing effects of these compounds. Overall, we presented a cell-based screening for NHEJ inhibitors, which could promote the discovery of novel radiosensitizers. Mol Cancer Ther; 17(2); 419-31. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Subject(s)
CRISPR-Cas Systems/genetics , Ouabain/therapeutic use , Penfluridol/therapeutic use , Humans , Ouabain/pharmacology , Penfluridol/pharmacology , Radiation-Sensitizing Agents , Transfection
11.
Sci Rep ; 7: 42928, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338004

ABSTRACT

The up-regulation of thioredoxin reductase-1 (TrxR1) is detected in more than half of gliomas, which is significantly associated with increased malignancy grade and recurrence rate. The biological functions of NADPH-dependent TrxR1 are mainly associated with reduced thioredoxin-1 (Trx1) which plays critical roles in cellular redox signaling and tumour radio-resistance. Our previous work has proved that TP53 induced glycolysis and apoptosis regulator (TIGAR) knockdown could notably radiosensitize glioma cells. However, whether TrxR1-overexpressing glioma cells could be re-radiosensitized by TIGAR silence is still far from clear. In the present study, TrxR1 was stably over-expressed in U-87MG and T98G glioma cells. Both in vitro and in vivo data demonstrated that the radiosensitivity of glioma cells was considerably diminished by TrxR1 overexpression. TIGAR abrogation was able to radiosensitize TrxR1-overexpressing gliomas by inhibiting IR-induced Trx1 nuclear transport. Post-radiotherapy, TIGAR low-expression predicted significant longer survival time for animals suffering from TrxR1-overexpessing xenografts, which suggested that TIGAR abrogation might be a promising strategy for radiosensitizing TrxR1-overexpressing glial tumours.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Thioredoxins/metabolism , Active Transport, Cell Nucleus/radiation effects , Animals , Apoptosis Regulatory Proteins , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , DNA Damage/radiation effects , Female , Glioma/mortality , Glioma/pathology , Glioma/radiotherapy , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , NADP/metabolism , Phosphoric Monoester Hydrolases , RNA Interference , RNA, Small Interfering/metabolism , Radiation Tolerance , Radiation, Ionizing , Reactive Oxygen Species/metabolism
12.
Oncotarget ; 8(2): 2457-2465, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27974693

ABSTRACT

Overexpression of epidermal growth factor receptor can be found in more than 80% of patients with locoregionally advanced nasopharyngeal carcinoma and is associated with shorter survival. In this work, we evaluated the feasibility of adding nimotuzumab to chemoradiation in locoregionally advanced nasopharyngeal carcinoma. Twenty-three patients with clinically staged T3-4 or any node-positive disease were enrolled. They were scheduled to receive one cycle of induction chemotherapy followed by intensity-modulated radiotherapy, weekly administration of nimotuzumab and concurrent chemotherapy. Results showed that all patients received a full course of radiotherapy, 19(82.6%)patients completed the scheduled neoadjuvant and concurrent chemotherapy, and 22(95.7%) patients received ≥6 weeks of nimotuzumab. During the period of concurrent chemoradiation and nimotuzumab, grade 3-4 toxicities occurred in 14(60.9%) patients: 8 (34.8%) had grade 3-4 oral mucositis, 6(26.1%) had grade 3 neutropenia, and 1(4.3%) had grade 3 dermatitis. No acne-like rash was observed. With a median follow-up of 24.1 months, the 2-year progression-free survival and overall survival were 83.5% and 95.0%, respectively. In conclusion, concurrent administration of chemoradiation and nimotuzumab was well-tolerated with good compliance. Preliminary clinical outcome data appear encouraging with favorable normal tissue toxicity results comparing with historical data of concurrent chemoradiation plus cetuximab.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Carcinoma/therapy , Chemoradiotherapy/methods , Induction Chemotherapy/methods , Nasopharyngeal Neoplasms/therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma/pathology , Feasibility Studies , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neoplasm Staging , Survival Analysis , Treatment Outcome , Young Adult
13.
Lung ; 194(1): 67-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26563331

ABSTRACT

PURPOSE: The present study was designed to investigate the effects of WP1066, a specific inhibitor of STAT3 signaling, on radiation-induced lung injury in mice. METHODS: C57BL/6J mice were subjected to a single thoracic irradiation of 15 Gy X-ray and WP1066 was administrated through intraperitoneal injection. The early and delayed treatment groups were treated with WP1066 during the first 2 weeks and the second 2 weeks, respectively. The therapeutic effects of WP1066 were evaluated by survival analysis, histological examination, and measurement of inflammatory parameters and collagen deposition. The activation of STAT3 pathway was also estimated by immunohistochemical staining and Western blotting. RESULTS: Delayed treatment of WP1066, but not early treatment, prolonged survival time and prevented the development of radiation pneumonitis and the subsequent lung fibrosis in mice. WP1066 treatment also significantly suppressed the activation of STAT3 signaling in the irradiated lung tissues. CONCLUSIONS: The activation of STAT3 pathway might play an important part in the pathogenesis of radiation-induced lung injury. The protective effects of delayed treatment of WP1066 suggested STAT3 signaling could be a therapeutic target for radiation pneumonitis.


Subject(s)
Lung/pathology , Pyridines/administration & dosage , RNA, Messenger/blood , Radiation Injuries, Experimental/drug therapy , Radiation Pneumonitis/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/administration & dosage , Animals , Drug Administration Schedule , Female , Fibrosis , Gene Expression/drug effects , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Interleukin-6/genetics , Lung/metabolism , Lung/radiation effects , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/pathology , Radiation Pneumonitis/blood , Radiation Pneumonitis/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
14.
Int J Biol Sci ; 11(12): 1458-68, 2015.
Article in English | MEDLINE | ID: mdl-26681925

ABSTRACT

BACKGROUND & AIMS: High doses of radiation induce severe DNA damage in intestinal epithelial cells, especially crypt cells, and cause intestinal injury, but the underlying molecular mechanisms remain unclear. Krüppel-like factor 5 (KLF5), a zinc finger-containing transcription factor, is induced by various stress stimuli and is involved in cell proliferation and survival. The role of KLF5 in radiation-induced intestinal injury was investigated here. METHODS: Wild type mice were treated with 8 or 15 Gy total body irradiation (TBI). KLF5 content and cellular localization in the small intestines of irradiated mice were detected by Western blot and immunohistochemical analysis. Mice with intestinal-specific knockdown of KLF5 (Vil-Cre; Klf5(fl/+) mice) were generated and their response to radiation was compared with controls. Morphological changes were determined by hematoxylin and eosin staining. Proliferation was examined by Ki67 immunostaining. The molecular response of the small intestine after KLF5 knockdown was investigated using microarrays. RESULTS: KLF5 expression correlated with the progression of intestinal damage. Decreased levels of KLF5 in the gut were associated with increased damage to the intestinal mucosa and reduced epithelial proliferation after TBI. Our microarray data disclosed that KLF5 knockdown down-regulated genes related to DNA damage repair pathways such as nucleotide excision repair, mismatch repair, non-homologous end joining and the Fanconi anemia pathway, which may suggest a novel function of KLF5. CONCLUSIONS: Our study illustrates that KLF5 may modulate DNA repair pathways to prevent intestinal injury induced by TBI. KLF5 signaling provides a novel field for identification of potential therapeutic targets for the treatment of radiation-induced intestinal damage.


Subject(s)
Cell Proliferation/physiology , DNA Damage , DNA Repair , Intestinal Mucosa/cytology , Intestine, Small/radiation effects , Kruppel-Like Transcription Factors/physiology , Animals , Down-Regulation , Intestine, Small/cytology , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL
15.
PLoS One ; 10(8): e0133728, 2015.
Article in English | MEDLINE | ID: mdl-26266801

ABSTRACT

OBJECTIVES: To compare the biological effects of 125I seeds continuous low-dose-rate (CLDR) radiation and 60Co γ-ray high-dose-rate (HDR) radiation on non-small cell lung cancer (NSCLC) cells. MATERIALS AND METHODS: A549, H1299 and BEAS-2B cells were exposed to 125I seeds CLDR radiation or 60Co γ-ray HDR radiation. The survival fraction was determined using a colony-forming assay. The cell cycle progression and apoptosis were detected by flow cytometry (FCM). The expression of the apoptosis-related proteins caspase-3, cleaved-caspase-3, PARP, cleaved-PARP, BAX and Bcl-2 were detected by western blot assay. RESULTS: After irradiation with 125I seeds CLDR radiation, there was a lower survival fraction, more pronounced cell cycle arrest (G1 arrest and G2/M arrest in A549 and H1299 cells, respectively) and a higher apoptotic ratio for A549 and H1299 cells than after 60Co γ-ray HDR radiation. Moreover, western blot assays revealed that 125I seeds CLDR radiation remarkably up-regulated the expression of Bax, cleaved-caspase-3 and cleaved-PARP proteins and down-regulated the expression of Bcl-2 proteins in A549 and H1299 cells compared with 60Co γ-ray HDR radiation. However, there was little change in the apoptotic ratio and expression of apoptosis-related proteins in normal BEAS-2B cells receiving the same treatment. CONCLUSIONS: 125I seeds CLDR radiation led to remarkable growth inhibition of A549 and H1299 cells compared with 60Co HDR γ-ray radiation; A549 cells were the most sensitive to radiation, followed by H1299 cells. In contrast, normal BEAS-2B cells were relatively radio-resistant. The imbalance of the Bcl-2/Bax ratio and the activation of caspase-3 and PARP proteins might play a key role in the anti-proliferative effects induced by 125I seeds CLDR radiation, although other possibilities have not been excluded and will be investigated in future studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Survival/radiation effects , Gamma Rays/therapeutic use , Neoplasm Proteins/biosynthesis , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/radiation effects , Cell Line, Tumor , Cobalt Radioisotopes/adverse effects , Cobalt Radioisotopes/therapeutic use , Dose-Response Relationship, Radiation , Flow Cytometry , Gamma Rays/adverse effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Iodine Radioisotopes/adverse effects , Iodine Radioisotopes/therapeutic use , Neoplasm Proteins/genetics
16.
Oncotarget ; 6(26): 22006-27, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26124081

ABSTRACT

Glioma stem cells (GSCs) exhibit stem cell properties and high resistance to radiotherapy. The main aim of our study was to determine the roles of ROS in radioresistance and stemness of GSCs. We found that microRNA (miR)-153 was down-regulated and its target gene nuclear factor-erythroid 2-related factor-2 (Nrf-2) was up-regulated in GSCs compared with that of non-GSCs glioma cells. The enhanced Nrf-2 expression increased glutathione peroxidase 1 (GPx1) transcription and decreased ROS level leading to radioresistance of GSCs. MiR-153 overexpression resulted in increased ROS production and radiosensitization of GSCs. Moreover, miR-153 overexpression led to decreased neurosphere formation capacity and stem cell marker expression, and induced differentiation through ROS-mediated activation of p38 MAPK in GSCs. Nrf-2 overexpression rescued the decreased stemness and radioresistance resulting from miR-153 overexpression in GSCs. In addition, miR-153 overexpression reduced tumorigenic capacity of GSCs and increased survival in mice bearing human GSCs. These findings demonstrated that miR-153 overexpression decreased radioresistance and stemness of GSCs through targeting Nrf-2/GPx1/ROS pathway.


Subject(s)
Brain Neoplasms/radiotherapy , Glioma/radiotherapy , Glutathione Peroxidase/metabolism , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/radiation effects , Reactive Oxygen Species/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/radiation effects , Down-Regulation , Female , Glioma/metabolism , Glioma/pathology , Glutathione Peroxidase/genetics , Humans , Mice , MicroRNAs/genetics , Middle Aged , NF-E2-Related Factor 2/genetics , Neoplastic Stem Cells/pathology , Radiation Tolerance/physiology , Signal Transduction , Transfection , Xenograft Model Antitumor Assays , Glutathione Peroxidase GPX1
17.
Int J Clin Exp Pathol ; 8(5): 4823-9, 2015.
Article in English | MEDLINE | ID: mdl-26191173

ABSTRACT

Our previous study proved that TP53-induced glycolysis and apoptosis regulator (TIGAR) abrogation is able to radiosensitize glioma cells. Whether TIGAR over-expression has radio-protective effect in human parotid gland cells is still unknown. In this study human parotid gland fibroblast Hs 917.T cells were transfected with pcDNA3.1-TIGAR, and clonogenic assay was performed to investigate the radiosensitivity of Hs 917.T cells over-expressing pcDNA3.1 or pcDNA3.1-TIGAR. Western blot was carried out to demonstrate the autophagy activity of cells being irradiated, and immunofluorescence assay was used to evaluate the DNA damage repair process of irradiated Hs 917.T cells. It was revealed that TIGAR over-expression could diminish the radiosensitivity of Hs 917.T cells, and the autophagy level induced by ionizing radiation (IR) was also decreased by TIGAR transfection. The mechanism might rely on TIGAR over-expression induced ROS scavenging and NADPH increasing. Using autophagy inhibitor, it was also elaborated that IR-induced autophagy in Hs 917.T cells was protective autophagy but not traumatic autophagy.


Subject(s)
Autophagy/genetics , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Parotid Gland/cytology , Radiation Tolerance/genetics , Apoptosis Regulatory Proteins , Cell Line , Fibroblasts/cytology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Parotid Gland/metabolism , Phosphoric Monoester Hydrolases , Reactive Oxygen Species/metabolism
18.
Oncol Lett ; 10(1): 473-478, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26171054

ABSTRACT

The present study aimed to investigate the radiosensitizing effects of tamoxifen (TAM), a non-steroidal anti-estrogen drug, in human glioma A172 and U251 cells in vitro. A colony-forming assay revealed that TAM enhances radiosensitivity in A172 and U251 cells. Treatment with TAM also increased the percentage of apoptotic cells subsequent to ionizing radiation, and increased the expression of apoptotic markers, including cleaved caspase-3 and poly(ADP-ribose) polymerase. Ionizing radiation induced G2/M phase arrest, which was alleviated within 24 h when the radiation-induced DNA damage was repaired. However, flow cytometry analysis revealed that TAM treatment delayed the recovery of cell cycle progression. Additional examination demonstrated that TAM-mediated protein kinase C-ι (PKC-ι) inhibition may lead to the activation of pro-apoptotic B-cell lymphoma 2-associated death promoter, and the dephosphorylation of cyclin-dependent kinase 7, resulting in increased cell apoptosis and sustained G2/M phase arrest following exposure to radiation. The present data indicate that the radiosensitizing effects of TAM on glioma cells are partly due to the inhibition of PKC-ι activity in vitro.

19.
World J Gastroenterol ; 21(8): 2336-42, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25741139

ABSTRACT

AIM: To determine the mechanism of the radiation-induced biological effects of 125I seeds on pancreatic carcinoma cells in vitro. METHODS: SW1990 and PANC-1 pancreatic cancer cell lines were cultured in DMEM in a suitable environment. Gray's model of iodine-125 (125I) seed irradiation was used. In vitro, exponential phase SW1990, and PANC-1 cells were exposed to 0, 2, 4, 6, and 8 Gy using 125I radioactive seeds, with an initial dose rate of 12.13 cGy/h. A clonogenic survival experiment was performed to observe the ability of the cells to maintain their clonogenic capacity and to form colonies. Cell-cycle and apoptosis analyses were conducted to detect the apoptosis percentage in the SW1990 and PANC-1 cells. DNA synthesis was measured via a tritiated thymidine (3H-TdR) incorporation experiment. After continuous low-dose-rate irradiation with 125I radioactive seeds, the survival fractions at 2 Gy (SF2), percentage apoptosis, and cell cycle phases of the SW1990 and PANC-1 pancreatic cancer cell lines were calculated and compared. RESULTS: The survival fractions of the PANC-1 and SW1990 cells irradiated with 125I seeds decreased exponentially as the dose increased. No significant difference in SF2 was observed between SW1990 and PANC-1 cells (0.766±0.063 vs 0.729±0.045, P<0.05). The 125I seeds induced a higher percentage of apoptosis than that observed in the control in both the SW1990 and PANC-1 cells. The rate of apoptosis increased with increasing radiation dosage. The percentage of apoptosis was slightly higher in the SW1990 cells than in the PANC-1 cells. Dose-dependent G2/M cell-cycle arrest was observed after 125I seed irradiation, with a peak value at 6 Gy. As the dose increased, the percentage of G2/M cell cycle arrest increased in both cell lines, whereas the rate of DNA incorporation decreased. In the 3H-TdR incorporation experiment, the dosimetry results of both the SW1990 and PANC-1 cells decreased as the radiation dose increased, with a minimum at 6 Gy. There were no significant differences in the dosimetry results of the two cell lines when they were exposed to the same dose of radiation. CONCLUSION: The pancreatic cancer cell-killing effects induced by 125I radioactive seeds mainly occurred via apoptosis and G2/M cell cycle arrest.


Subject(s)
Brachytherapy/methods , Carcinoma, Pancreatic Ductal/radiotherapy , Iodine Radioisotopes , Pancreatic Neoplasms/radiotherapy , Apoptosis/radiation effects , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , G2 Phase Cell Cycle Checkpoints/radiation effects , Humans , Pancreatic Neoplasms/pathology
20.
Gene ; 560(2): 237-44, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25680556

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of cancer related deaths worldwide. RNA helicases have been widely implicated in various types of cancer development. DDX46 belongs to the DEAD box family of RNA helicases, which are involved in the regulation of secondary RNA structures. The expression pattern of DDX46 in cancer tissues and the role of DDX46 in CRC progression have not been determined. In this study, we detected DDX46 protein expression in human CRC and adjacent tissues using immunohistochemistry. Our results showed that 87.04% of the columnar adenocarcinoma cases displayed high levels of focal nuclear DDX46 staining, and DDX46 protein expression was strongly increased in CRC tissues compared to adjacent tissues. Next, DDX46 RNAi lentivirus (DDX46-RNAi-LV) was used to silence the expression of DDX46 in the human colon carcinoma cells. Cells treated with the DDX46-RNAi-LV exhibited markedly reduced cell proliferation assessed by the MTT assay and visualized colony formation. Moreover, DDX46 silencing resulted in apoptotic induction via increased expression of cleaved caspase-3 and PARP. These results indicate that DDX46 is critical for CRC cell proliferation and is a potential therapeutic target for CRC treatment.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis , Colorectal Neoplasms/metabolism , DEAD-box RNA Helicases/genetics , Lentivirus/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Proliferation , Cell Survival , DEAD-box RNA Helicases/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , Humans , Male , Middle Aged , Ribonucleoprotein, U2 Small Nuclear/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...