Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 9(1): 291, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37558683

ABSTRACT

Radiotherapy resistance is an important and urgent challenge in the clinical management of esophageal squamous carcinoma (ESCC). However, the factors mediating the ESCC resistance to radiotherapy and its underlying molecular mechanisms are not fully clarified. Our previous studies have demonstrated the critical role of DNA polymerase iota (POLI) in ESCC development and progression, here, we aimed to investigate the involvement of POLI in ESCC radiotherapy resistance and elucidate the underlying molecular mechanism. We found that highly expressed POLI was correlated with shorter overall survival of ESCC patients received radiotherapy. Down-regulation of POLI sensitized ESCC to IR, prolonged γH2AX foci in nuclei and comet tails after IR. HR but not NHEJ repair is inhibited in POLI-deficient ESCC cells. POLI stabilizes RAD51 protein via competitively binding with and blocking the interaction between RAD51 and E3 ligase XIAP and XIAP-mediated ubiquitination. Furthermore, loss of POLI leads to the activation of GAS signaling. Our findings provide novel insight into the role of POLI in the development of radioresistance mediated by stabilizing RAD51 protein in ESCC.

2.
Exploration (Beijing) ; 3(2): 20220119, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37324033

ABSTRACT

Ionizing radiation (IR) poses a growing threat to human health, and thus ideal radioprotectors with high efficacy and low toxicity still receive widespread attention in radiation medicine. Despite significant progress made in conventional radioprotectants, high toxicity, and low bioavailability still discourage their application. Fortunately, the rapidly evolving nanomaterial technology furnishes reliable tools to address these bottlenecks, opening up the cutting-edge nano-radioprotective medicine, among which the intrinsic nano-radioprotectants characterized by high efficacy, low toxicity, and prolonged blood retention duration, represent the most extensively studied class in this area. Herein, we made the systematic review on this topic, and discussed more specific types of radioprotective nanomaterials and more general clusters of the extensive nano-radioprotectants. In this review, we mainly focused on the development, design innovations, applications, challenges, and prospects of the intrinsic antiradiation nanomedicines, and presented a comprehensive overview, in-depth analysis as well as an updated understanding of the latest advances in this topic. We hope that this review will promote the interdisciplinarity across radiation medicine and nanotechnology and stimulate further valuable studies in this promising field.

4.
Cells ; 11(10)2022 05 16.
Article in English | MEDLINE | ID: mdl-35626687

ABSTRACT

Renal cell carcinoma (RCC) is one of the most aggressive urological malignancies and has a poor prognosis, especially in patients with metastasis. Although RCC is traditionally considered to be radioresistant, radiotherapy (RT) is still a common treatment for palliative management of metastatic RCC. Novel approaches are urgently needed to overcome radioresistance of RCC. Black phosphorus quantum dots (BPQDs) have recently received great attention due to their unique physicochemical properties and good biocompatibility. In the present study, we found that BPQDs enhance ionizing radiation (IR)-induced apoptotic cell death of RCC cells. BPQDs treatment significantly increases IR-induced DNA double-strand breaks (DSBs), as indicated by the neutral comet assay and the DSBs biomarkers γH2AX and 53BP1. Mechanistically, BPQDs can interact with purified DNA-protein kinase catalytic subunit (DNA-PKcs) and promote its kinase activity in vitro. BPQDs impair the autophosphorylation of DNA-PKcs at S2056, and this site phosphorylation is essential for efficient DNA DSBs repair and the release of DNA-PKcs from the damage sites. Consistent with this, BPQDs suppress nonhomologous end-joining (NHEJ) repair and lead to sustained high levels of autophosphorylated DNA-PKcs on the damaged sites. Moreover, animal experiments indicate that the combined approach with both BPQDs and IR displays better efficacy than monotreatment. These findings demonstrate that BPQDs have potential applications in radiosensitizing RCC cells.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Quantum Dots , Animals , Carcinoma, Renal Cell/radiotherapy , DNA/metabolism , DNA Repair , Humans , Kidney Neoplasms/radiotherapy , Phosphorus , Polynucleotide 5'-Hydroxyl-Kinase/metabolism , Radiation Tolerance
5.
Oncogene ; 41(4): 489-501, 2022 01.
Article in English | MEDLINE | ID: mdl-34775484

ABSTRACT

Chromosomal instability (CIN) is a driving force for cancer development. The most common causes of CIN include the dysregulation of the spindle assembly checkpoint (SAC), which is a surveillance mechanism that prevents premature chromosome separation during mitosis by targeting anaphase-promoting complex/cyclosome (APC/C). DAB2IP is frequently silenced in advanced prostate cancer (PCa) and is associated with aggressive phenotypes of PCa. Our previous study showed that DAB2IP activates PLK1 and functions in mitotic regulation. Here, we report the novel mitotic phosphorylation of DAB2IP by Cdks, which mediates DAB2IP's interaction with PLK1 and the activation of the PLK1-Mps1 pathway. DAB2IP interacts with Cdc20 in a phosphorylation-independent manner. However, the phosphorylation of DAB2IP inhibits the ubiquitylation of Cdc20 in response to SAC, and blocks the premature release of the APC/C-MCC. The PLK1-Mps1 pathway plays an important role in mitotic checkpoint complex (MCC) assembly. It is likely that DAB2IP acts as a scaffold to aid PLK1-Mps1 in targeting Cdc20. Depletion or loss of the Cdks-mediated phosphorylation of DAB2IP destabilizes the MCC, impairs the SAC, and increases chromosome missegregation and subsequent CIN, thus contributing to tumorigenesis. Collectively, these results demonstrate the mechanism of DAB2IP in SAC regulation and provide a rationale for targeting the SAC to cause lethal CIN against DAB2IP-deficient aggressive PCa, which exhibits a weak SAC.


Subject(s)
Cell Cycle Checkpoints/genetics , Chromosomal Instability/genetics , Mitosis/genetics , Oncogenes/genetics , Spindle Apparatus/metabolism , Humans , Phosphorylation , Signal Transduction , Transfection
6.
Cell Cycle ; 20(2): 211-224, 2021 01.
Article in English | MEDLINE | ID: mdl-33404279

ABSTRACT

Combining targeted therapeutic agents is an attractive cancer treatment strategy associated with high efficacy and low toxicity. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is an essential factor in DNA damage repair. Studies from us and others have revealed that DNA-PKcs also plays an important role in normal mitosis progression. Histone deacetylase (HDACs) inhibitors commonly lead to mitotic aberration and have been approved for treating various cancers in the clinic. We showed that DNA-PKcs depletion or kinase activity inhibition increases cancer cells' sensitivity to HDACs inhibitors in vitro and in vivo. DNA-PKcs deficiency significantly enhances HDACs inhibitors (HDACi)-induced mitotic arrest and is followed by apoptotic cell death. Mechanistically, we found that DNA-PKcs binds to HDAC6 and facilitates its acetylase activity. HDACi is more likely to impair HDAC6-induced deacetylation of HSP90 and abrogate HSP90's chaperone function on Aurora A, a critical mitotic kinase that regulates centrosome separation and mitotic spindle assembly in DNA-PKcs-deficient cells. Our current work indicates crosstalk between DNA-PKcs and HDACs signaling pathways, and highlights that the combined targeting of DNA-PKcs and HDACs can be used in cancer therapy. Abbreviations: DNA-PKcs, DNA-dependent protein kinase catalytic subunit, HDACs, Histone deacetylases, DSBs, DNA double-strand breaks, ATM, ataxia telangiectasia mutated, ATR, ATM-Rad3-related.


Subject(s)
DNA-Activated Protein Kinase/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6/metabolism , Molecular Chaperones/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus Division/genetics , Cell Nucleus Division/physiology , DNA Damage/genetics , DNA Repair/genetics , Histone Deacetylase 6/genetics , Humans , Tumor Suppressor Proteins/metabolism
7.
Oncogenesis ; 10(1): 8, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33431808

ABSTRACT

The PI3K/AKT/mTOR signaling pathway is constitutively active in PTEN-deficient cancer cells, and its targeted inhibition has significant anti-tumor effects. However, the efficacy of targeted therapies is often limited due to drug resistance. The relevant signaling pathways in PTEN-deficient cancer cells treated with the PI3K/mTOR inhibitor BEZ235 were screened using a phosphokinase array, and further validated following treatment with multiple PI3K/AKT/mTOR inhibitors or AKT knockdown. The correlation between PTEN expression levels and STAT3 kinase phosphorylation in the tissue microarrays of gastric cancer patients was analyzed by immunohistochemistry. Cell proliferation and clonogenic assays were performed on the suitably treated PTEN-deficient cancer cells. Cytokine arrays, small molecule inhibition and knockdown assays were performed to identify related factors. PTEN-deficient tumor xenografts were established in nude mice that were treated with PI3K/AKT/mTOR and/or STAT3 inhibitors. PTEN deficiency was positively correlated with low STAT3 activity. PI3K/mTOR inhibitors increased the expression and secretion of macrophage migration inhibitory factor (MIF) and activated the JAK1/STAT3 signaling pathway. Both cancer cells and in vivo tumor xenografts showed that the combined inhibition of PI3K/AKT/mTOR and STAT3 activity enhanced the inhibitory effect of BEZ235 on the proliferation of PTEN-deficient cancer cells. Our findings provide a scientific basis for a novel treatment strategy in cancer patients with PTEN deficiency.

8.
Toxicol Appl Pharmacol ; 387: 114855, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31830491

ABSTRACT

Vanillin is a natural compound endowed with antioxidant and anti-mutagenic properties. We previously identified the vanillin derivative VND3207 with strong radio-protective and antioxidant effects and found that VND3207 confers survival benefit and protection against radiation-induced intestinal injury (RIII) in mice. We also observed that VND3207 treatment enhanced the expression level of the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs) in human lymphoblastoid cells with or without γ-irradiation. DNA-PKcs is a critical component of DNA double strand break repair pathway and also regulates mitotic progression by stabilizing spindle formation and preventing mitotic catastrophe in response to DNA damage. In the present study, we found that VND3207 protected intestinal epithelial cells in vitro against ionizing radiation by promoting cell proliferation and inhibiting cell apoptosis. In addition, VND3207 promoted DNA-PKcs activity by increasing autophosphorylation at S2056 site. Consistent with this, VND3207 significantly decreased the number of γH2AX foci and mitotic catastrophe after radiation. DNA-PKcs deficiency abolished these VND3207 radio-protective effects, indicating that DNA-PKcs activation is essential for VND3207 activity. In conclusion, VND3207 promoted intestinal repair following radiation injury by regulating the DNA-PKcs pathway.


Subject(s)
Benzaldehydes/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , DNA-Activated Protein Kinase/metabolism , Intestinal Mucosa/drug effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA-Activated Protein Kinase/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Gamma Rays/adverse effects , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Loss of Function Mutation , Male , Mice , Phosphorylation/drug effects , Radiation Injuries, Experimental/pathology , Radiation-Protective Agents/therapeutic use
9.
Free Radic Biol Med ; 145: 223-236, 2019 12.
Article in English | MEDLINE | ID: mdl-31580946

ABSTRACT

The intestine is a highly radiosensitive tissue that is susceptible to structural and functional damage due to systemic as well as localized radiation exposure. Unfortunately, no effective prophylactic or therapeutic agents are available at present to manage radiation-induced intestinal injuries. We observed that the vanillin derivative VND3207 improved the survival of lethally irradiated mice by promoting intestinal regeneration and increasing the number of surviving crypts. Pre-treatment with VND3207 significantly increased the number of Lgr5+ intestinal stem cells (ISCs) and their daughter cells, the transient Ki67+ proliferating cells. Mechanistically, VND3207 decreased oxidative DNA damage and lipid peroxidation and maintained endogenous antioxidant status by increasing the level of superoxide dismutase and total antioxidant capacity. In addition, VND3207 maintained appropriate levels of activated p53 that triggered cell cycle arrest but were not sufficient to induce NOXA-mediated apoptosis, thus ensuring DNA damage repair in the irradiated small intestinal crypt cells. Furthermore, VND3207 treatment restores the intestinal bacterial flora structures altered by TBI exposure. In conclusion, VND3207 promoted intestinal repair following radiation injury by reducing reactive oxygen species-induced DNA damage and modulating appropriate levels of activated p53 in intestinal epithelial cells.


Subject(s)
Benzaldehydes/pharmacology , Gastrointestinal Microbiome/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, G-Protein-Coupled/genetics , Tumor Suppressor Protein p53/genetics , Animals , Antioxidants/pharmacology , Benzaldehydes/chemistry , Cell Lineage/drug effects , Cell Lineage/radiation effects , Gastrointestinal Microbiome/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Intestines/drug effects , Mice , Oxidative Stress/drug effects , Radiation Exposure/adverse effects , Radiation Injuries/genetics , Radiation Injuries/prevention & control , Radiation Tolerance/genetics , Signal Transduction/radiation effects , Stem Cells/drug effects
10.
Nanotoxicology ; 13(10): 1409-1421, 2019 12.
Article in English | MEDLINE | ID: mdl-31589482

ABSTRACT

Graphene quantum dots (GQDs) have gained significant attention in various biomedical applications. The physicochemical properties of these nanoparticles, including toxic effects, are largely determined by their surface modifications. Previous studies have demonstrated high in vitro cytotoxicity of the hydroxylated GQDs (OH-GQDs). The focus of this study was on the intestinal toxicity of OH-GQDs. Briefly, C57BL/6J mice were given daily oral gavage of 0.05, 0.5 or 5 mg/kg OH-GQD for 7 days, and the indices of intestinal damage were evaluated. Higher doses of the OH-GQDs caused significant intestinal injuries, such as enhanced intestinal permeability, shortened villi and crypt loss. The number of Lgr5+ intestinal stem cells also decreased dramatically upon OH-GQDs exposure, which also inhibited the Ki67+ proliferative progenitor cells. In addition, an increased number of crypt cells harboring the oxidized DNA base 8-OHdG and γH2AX foci were also detected in the intestines of OH-GQD-treated mice. Mechanistically, the OH-GQDs up-regulated both total and phosphorylated p53. Consistent with this, the average number of TUNEL+ and cleaved caspase-3+ apoptotic intestinal epithelial cells were significantly increased after OH-GQDs treatment. Finally, a 3-dimensional organoid culture was established using isolated crypts, and OH-GQDs treatment significantly reduced the size of the surviving intestinal organoids. Taken together, the intestinal toxicity of the OH-GQDs should be taken into account during biomedical applications.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Graphite/toxicity , Intestinal Mucosa/drug effects , Quantum Dots/toxicity , Stem Cells/drug effects , Administration, Oral , Animals , Apoptosis/genetics , Cell Proliferation/genetics , DNA Damage , Graphite/chemistry , Hydroxylation , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Permeability , Quantum Dots/chemistry , Stem Cells/pathology , Surface Properties , Tumor Suppressor Protein p53/genetics
11.
Cancer Sci ; 109(12): 3783-3793, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30281878

ABSTRACT

The p53-inducible gene 3 (PIG3) is one of the p53-induced genes at the onset of apoptosis, which plays an important role in cell apoptosis and DNA damage response. Our previous study reported an oncogenic role of PIG3 associated with tumor progression and metastasis in non-small cell lung cancer (NSCLC). In this study, we further analyzed PIG3 mRNA expression in 504 lung adenocarcinoma (LUAD) and 501 lung squamous cell carcinoma (LUSC) tissues from The Cancer Genome Atlas database and we found that PIG3 expression was significantly higher in LUAD with lymph node metastasis than those without, while no difference was observed between samples with and without lymph node metastasis in LUSC. Gain and loss of function experiments were performed to confirm the metastatic role of PIG3 in vitro and to explore the mechanism involved in its oncogenic role in NSCLC metastasis. The results showed that PIG3 knockdown significantly inhibited the migration and invasion ability of NSCLC cells, and decreased paxillin, phospho-focal adhesion kinase (FAK) and phospho-Src kinase expression, while its overexpression resulted in the opposite effects. Blocking FAK with its inhibitor reverses PIG3 overexpression-induced cell motility in NSCLC cells, indicating that PIG3 increased cell metastasis through the FAK/Src/paxillin pathway. Furthermore, PIG3 silencing sensitized NSCLC cells to FAK inhibitor. In conclusion, our data revealed a role for PIG3 in inducing LUAD metastasis, and its role as a new FAK regulator, suggesting that it could be considered as a novel prognostic biomarker or therapeutic target in the treatment of LUAD metastasis.


Subject(s)
Adenocarcinoma of Lung/genetics , Carcinoma, Squamous Cell/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction , Up-Regulation , A549 Cells , Adenocarcinoma of Lung/metabolism , Adult , Aged , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Lymphatic Metastasis , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism
12.
Toxicol Appl Pharmacol ; 348: 76-84, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29679654

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the most common form of esophageal cancer in China. Since chemotherapy is the standard clinical intervention for advanced ESCC, the development of highly effective and minimal/non-toxic drugs is essential to improve the clinical outcome and prognosis of the patients. A novel derivative of vanillin, 6-bromine-5-hydroxy-4-methoxybenzaldehyde (BVAN08), has been recently reported to activate different cell death pathways in cancer cells. In this study, we demonstrate that BVAN08 exhibits a potent anti-proliferation effect on ESCC cells (TE-1 and ECA-109) by inhibiting the expression of PLK1, an important mitotic kinase. Consistent with this, BVAN08 induces mitotic arrest and chromosomal misalignment in ESCC cells. The disruption of microtubule nucleation around centrosomes is also observed in BVAN08 treated ESCC cells. Furthermore, BVAN08 enhances radio-sensitivity of ESCC cells by prolonging DNA damage repair. These findings underscore the potential value of BVAN08 in cancer therapeutics and demonstrate the underlying mechanism by which BVAN08 induces mitotic catastrophe and enhances radio-sensitivity in ESCC cells.


Subject(s)
Antineoplastic Agents/pharmacology , Benzaldehydes/pharmacology , Carcinoma, Squamous Cell/therapy , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Chemoradiotherapy , Esophageal Neoplasms/therapy , Mitosis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Radiation Tolerance/drug effects , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Centrosome/drug effects , Centrosome/pathology , DNA Damage , DNA Repair/drug effects , Dose-Response Relationship, Drug , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Microtubules/drug effects , Microtubules/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Time Factors , Polo-Like Kinase 1
13.
Toxicol Sci ; 164(1): 339-352, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29669094

ABSTRACT

Graphene quantum dots (GQDs) have attracted significant interests due to their unique chemical and physical properties. In this study, we investigated the potential effects of hydroxyl-modified GQDs (OH-GQDs) on the human esophageal epithelial cell line HET-1A. Our data revealed significant cytotoxicity of OH-GQDs which decreased the viability of HET-1A in a dose and time-dependent manner. The moderate concentration (25 or 50 µg/ml) of OH-GQDs significantly blocked HET-1A cells in G0/G1 cell cycle phase. An increased percentage of γH2AX-positive and genomically unstable cells were also detected in cells treated with different doses of OH-GQDs (25, 50, and 100 µg/ml). Microarray data revealed that OH-GQDs treatment down-regulated genes related to DNA damage repair, cell cycle regulation and cytoskeleton signal pathways indicating a novel role of OH-GQDs. Consistent with the microarray data, OH-GQDs disrupted microtubule structure and inhibited microtubule regrowth around centrosomes in HET-1A cells. In conclusion, our findings provide important evidence for considering the application of OH-GQDs in biomedical fields.


Subject(s)
DNA Damage , Epithelial Cells/drug effects , Esophagus/drug effects , Graphite/toxicity , Microtubules/drug effects , Quantum Dots/toxicity , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA Repair/genetics , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/pathology , Esophagus/pathology , Gene Expression Regulation/drug effects , Graphite/chemistry , Humans , Hydroxylation , Microtubules/ultrastructure , Quantum Dots/chemistry , Time Factors
14.
Nucleic Acids Res ; 46(4): 1847-1859, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29309644

ABSTRACT

The DNA-dependent protein kinase (DNA-PK), consisting of the DNA binding Ku70/80 heterodimer and the catalytic subunit DNA-PKcs, has been well characterized in the non-homologous end-joining mechanism for DNA double strand break (DSB) repair and radiation resistance. Besides playing a role in DSB repair, DNA-PKcs is required for the cellular response to replication stress and participates in the ATR-Chk1 signaling pathway. However, the mechanism through which DNA-PKcs is recruited to stalled replication forks is still unclear. Here, we report that the apoptosis mediator p53-induced protein with a death domain (PIDD) is required to promote DNA-PKcs activity in response to replication stress. PIDD is known to interact with PCNA upon UV-induced replication stress. Our results demonstrate that PIDD is required to recruit DNA-PKcs to stalled replication forks through direct binding to DNA-PKcs at the N' terminal region. Disruption of the interaction between DNA-PKcs and PIDD not only compromises the ATR association and regulation of DNA-PKcs, but also the ATR signaling pathway, intra-S-phase checkpoint and cellular resistance to replication stress. Taken together, our results indicate that PIDD, but not the Ku heterodimer, mediates the DNA-PKcs activity at stalled replication forks and facilitates the ATR signaling pathway in the cellular response to replication stress.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Replication , DNA-Activated Protein Kinase/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Cricetinae , DNA-Activated Protein Kinase/chemistry , Humans , Ku Autoantigen/physiology , Nuclear Proteins/chemistry , S Phase Cell Cycle Checkpoints , Signal Transduction , Stress, Physiological , Ultraviolet Rays
15.
Cancer Sci ; 108(12): 2503-2510, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28905458

ABSTRACT

An aberrantly elevated expression of DNA polymerase ι (Pol ι) is significantly associated with poor prognosis of patients with esophageal squamous cell carcinoma (ESCC), yet the mechanisms behind this phenomenon remain obscure. Based on the RNA-Seq transcriptome and real-time PCR analysis, we identified ETS-1 as a candidate gene involved in Pol ι-mediated progression of ESCC. Wound-healing and transwell assay indicated that downregulation of ETS-1 attenuates Pol ι-mediated invasiveness of ESCC. Signaling pathway analysis showed that Pol ι enhances ETS-1 phosphorylation at threonine-38 through the Erk signaling pathway in ESCC cells. Kaplan-Meier analysis, based on 93 clinical tissue samples, revealed that ETS-1 phosphorylation at threonine-38 is associated with poor prognosis of ESCC patients. The present study thus demonstrates that phosphorylation of ETS-1 is a critical event in the Pol ι-induced invasion and metastasis of ESCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , DNA-Directed DNA Polymerase/metabolism , Esophageal Neoplasms/pathology , Proto-Oncogene Protein c-ets-1/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Movement , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma , Humans , Kaplan-Meier Estimate , Neoplasm Invasiveness/pathology , Phosphorylation , DNA Polymerase iota
16.
Int J Med Sci ; 14(5): 452-461, 2017.
Article in English | MEDLINE | ID: mdl-28539821

ABSTRACT

Objectives: 4E-BP1 is a family member of eIF4E binding proteins (4E-BPs) which act as the suppressors of cap-dependent translation of RNA via competitively associating with cap-bound eIF4E. RNA translation regulation is an important manner to control the cellular responses to a series of stress conditions such as ionizing radiation (IR)-induced DNA damage response and cell cycle controlling. This study aimed to determine the mechanism of 4E-BP1 stabilization and its potential downstream target(s) in the response to IR. Methods: PI3Ks kinase inhibitors were used to determine the signaling control of 4E-BP1 phosphorylation and protein stability. shRNA strategy was employed to silence the expression of 4E-BP1 in HeLa and HepG2 cells, and determine its effect on the irradiation-induced CHK2 phosphorylation. The protein degradation/stability was investigated by western blotting on the condition of blocking novel protein synthesis by cycloheximide (CHX). Results: The phosphorylation of 4E-BP1 at Thr37/46 was significantly increased in both HepG2 and HeLa cells by ionizing radiation. Depression of 4E-BP1 by shRNA strategy resulted in an incomplete G2 arrest at the early stage of 2 hours post-irradiation, as well as a higher accumulation of mitotic cells at 10 and 12 hours post-irradiation as compared to the control cells. Consistently, the CHK2 phosphorylation at Thr68 induced by IR was also attenuated by silencing 4E-BP1 expression. Both PI3K and DNA-PKcs kinase inhibitors significantly decreased the protein level of 4E-BP1, which was associated with the accelerated degradation mediated by ubiquitination-proteasome pathway. Conclusion: PI3K kinase activity is necessary for maintaining 4E-BP1 stability. Our results also suggest 4E-BP1 a novel biological role of regulating cell cycle G2 checkpoint in responding to IR stress in association with controlling CHK2 phosphorylation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Checkpoint Kinase 2/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/genetics , Protein Biosynthesis/genetics , Cell Cycle Proteins , G2 Phase Cell Cycle Checkpoints/radiation effects , Gene Expression Regulation/radiation effects , HeLa Cells , Hep G2 Cells , Humans , Phosphorylation/radiation effects , Protein Biosynthesis/radiation effects , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , Radiation, Ionizing , Signal Transduction/genetics , Signal Transduction/radiation effects
17.
J Exp Clin Cancer Res ; 36(1): 39, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28259183

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most commonly diagnosed type of lung cancer that is associated with poor prognosis. In this study we explored the potential role of p53-induced gene 3 (PIG3) in the progression of NSCLC. METHODS: Immunohistochemistry was used to determine the expression levels of PIG3 in 201 NSCLC patients. We performed in vitro studies and silenced endogenous PIG3 by using specific siRNAs that specific target PIG3. Immunofluorescent staining was performed to determine the effect of PIG3 on mitotic progression in NSCLC cells. The growth rates of microtubules were determined by microtubule nucleation analysis. Cell proliferation and chemosensitivity were analyzed by CCK8 assays. Annexin V staining and ß-galactosidase activity analysis were used to evaluate PIG3 deficiency-related apoptosis and senescence, respectively. RESULTS: PIG3 expression levels negatively correlated with overall survival and disease-free survival of NSCLC patients. Knock down of PIG3 resulted in repressed proliferation of NSCLC cells and increased aberrant mitosis, which included misaligning and lagging chromosomes, and bi- or multi-nucleated giant cells. In addition, PIG3 contributed to mitotic spindle assembly by promoting microtubule growth. Furthermore, loss of PIG3 sensitized NSCLC cells to docetaxel by enhancing docetaxel-induced apoptosis and senescence. CONCLUSIONS: Our results indicate that PIG3 promotes NSCLC progression and therefore suggest that PIG3 may be a potential prognostic biomarker and novel therapeutic target for the treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/genetics , Mitosis , Proto-Oncogene Proteins/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Disease-Free Survival , Docetaxel , Female , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Prognosis , Proto-Oncogene Proteins/genetics , Survival Analysis , Taxoids/pharmacology , Young Adult
18.
Oncotarget ; 7(38): 62340-62351, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27694690

ABSTRACT

Radiotherapy is promising and effective for treating prostate cancer but the addition of a tumor cell radiosensitizer would improve therapeutic outcomes. PC-1/PrLZ, a TPD52 protein family member is frequently upregulated in advanced prostate cancer cells and may be a biomarker of aggressive prostate cancer. Therefore, we investigated the potential role of PC-1/PrLZ for increasing radioresistance in human prostate cancer cell lines. Growth curves and survival assays after g-ray irradiation confirmed that depletion of endogenous PC-1/PrLZ significantly increased prostate cancer cell radiosensitivity. Irradiation (IR) increased PC-1/PrLZ expression in a dose- and time-dependent manner and increased radiosensitivity in PC-1/PrLZ-suppressed cells was partially due to decreased DNA double strand break (DBS) repair which was measured with comet and gH2AX foci assays. Furthermore, depletion of PC-1/PrLZ impaired the IR-induced G2/M checkpoint, which has been reported to be correlate with radioresistance in cancer cells. PC-1/PrLZ-deficient cells exhibited higher level of autophagy when compared with control cells. Thus, specific inhibition of PC-1/PrLZ might provide a novel therapeutic strategy for radiosensitizing prostate cancer cells.


Subject(s)
Autophagy/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Neoplasm Proteins/radiation effects , Prostatic Neoplasms/radiotherapy , Radiation Tolerance , Cell Line, Tumor , Dose-Response Relationship, Radiation , G2 Phase Cell Cycle Checkpoints/radiation effects , Gamma Rays , Histones/metabolism , Histones/radiation effects , Humans , Male , Microscopy, Fluorescence , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Prostate/cytology , Prostate/radiation effects , Prostatic Neoplasms/pathology , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation
19.
Nucleic Acids Res ; 44(18): 8842-8854, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27568005

ABSTRACT

Defects in kinetochore-microtubule (KT-MT) attachment and the spindle assembly checkpoint (SAC) during cell division are strongly associated with chromosomal instability (CIN). CIN has been linked to carcinogenesis, metastasis, poor prognosis and resistance to cancer therapy. We previously reported that the DAB2IP is a tumor suppressor, and that loss of DAB2IP is often detected in advanced prostate cancer (PCa) and is indicative of poor prognosis. Here, we report that the loss of DAB2IP results in impaired KT-MT attachment, compromised SAC and aberrant chromosomal segregation. We discovered that DAB2IP directly interacts with Plk1 and its loss inhibits Plk1 kinase activity, thereby impairing Plk1-mediated BubR1 phosphorylation. Loss of DAB2IP decreases the localization of BubR1 at the kinetochore during mitosis progression. In addition, the reconstitution of DAB2IP enhances the sensitivity of PCa cells to microtubule stabilizing drugs (paclitaxel, docetaxel) and Plk1 inhibitor (BI2536). Our findings demonstrate a novel function of DAB2IP in the maintenance of KT-MT structure and SAC regulation during mitosis which is essential for chromosomal stability.


Subject(s)
Cell Cycle Checkpoints , Chromosomal Instability , Kinetochores/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromosome Aberrations , Chromosome Segregation , Gene Knockout Techniques , Humans , Mice , Mitosis/drug effects , Mitosis/genetics , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA Interference , Tubulin Modulators/pharmacology , ras GTPase-Activating Proteins/genetics , Polo-Like Kinase 1
20.
Sci Rep ; 6: 30165, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417393

ABSTRACT

Radiation-induced bystander effect (RIBE) describes a set of biological effects in non-targeted cells that receive bystander signals from the irradiated cells. RIBE brings potential hazards to adjacent normal tissues in radiotherapy, and imparts a higher risk than previously thought. Excessive release of some substances from irradiated cells into extracellular microenvironment has a deleterious effect. For example, cytokines and reactive oxygen species have been confirmed to be involved in RIBE process via extracellular medium or gap junctions. However, RIBE-mediating signals and intercellular communication pathways are incompletely characterized. Here, we first identified a set of differentially expressed miRNAs in the exosomes collected from 2 Gy irradiated human bronchial epithelial BEP2D cells, from which miR-7-5p was found to induce autophagy in recipient cells. This exosome-mediated autophagy was significantly attenuated by miR-7-5p inhibitor. Moreover, our data demonstrated that autophagy induced by exosomal miR-7-5p was associated with EGFR/Akt/mTOR signaling pathway. Together, our results support the involvement of secretive exosomes in propagation of RIBE signals to bystander cells. The exosomes-containing miR-7-5p is a crucial mediator of bystander autophagy.


Subject(s)
Autophagy/physiology , Bronchi/metabolism , Bystander Effect/physiology , Epithelial Cells/metabolism , Exosomes/metabolism , MicroRNAs/metabolism , Cell Line , Humans , Radiation, Ionizing , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...