Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Pathology ; 52(4): 421-430, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32317175

ABSTRACT

Checkpoint kinase 2 (CHK2) is a serine-threonine kinase with a role in DNA repair, cell cycle arrest or apoptosis in response to DNA damage. Both reduced and increased CHK2 expression has been described in different tumour types with impact on patient prognosis. To evaluate prevalence and significance of altered CHK2 expression in prostate cancer, a tissue microarray containing 17,747 tumours was analysed by immunohistochemistry. Nuclear CHK2 immunostaining was absent or weak in benign prostate epithelium but often more prominent in cancers. CHK2 immunostaining was considered weak in 38.8%, moderate in 33.6% and strong in 11.2% of prostate cancers. High CHK2 expression was strongly associated with TMPRSS2:ERG fusions (p<0.0001). Subgroup analysis of ERG positive and negative cancers revealed that high CHK2 staining was significantly linked to advanced tumour stage, high Gleason score, positive nodal status, positive surgical margin, high preoperative PSA (p<0.0001 each) and early prostate-specific antigen (PSA) recurrence (p=0.0001) in the subset of ERG negative cancers, while most of these associations were absent in ERG positive cancers. In ERG negative cancers, high CHK2 expression was an independent predictor of patient prognosis, even if parameters were included that were only available postoperatively. High CHK2 expression was also linked to presence of chromosomal deletions, high level of androgen receptor expression, positive p53 immunostaining, and high Ki-67 labelling index. These provide further in vivo evidence for previously described functional interactions. In summary, high CHK2 expression is linked to adverse tumour features and independently predicts early biochemical recurrence in ERG negative prostate cancer. CHK2 measurement, either alone or in combination, might be of clinical utility in this prostate cancer subgroup.


Subject(s)
Biomarkers, Tumor/metabolism , Checkpoint Kinase 2/biosynthesis , Prostatic Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/analysis , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Retrospective Studies , Transcriptional Regulator ERG
2.
Chemosphere ; 238: 124586, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31442775

ABSTRACT

Cyantraniliprole can effectively control lepidopteran pests and has been used all over the world. In general, the risk of cyantraniliprole seems low for fish, but the toxicity selectivity among different fish species was not clear. Here the acute toxicity and chronic effects of cyantraniliprole to juvenile tilapia (Oreochromis mossambicus) were assessed. The results showed that 96 h LC50 of cyantraniliprole to tilapia was 38.0 mg/L. After exposed for 28 days, specific growth rates of the blank control, solution control, and the treatments of 0.037, 0.37 and 3.7 mg/L of cyantraniliprole were 1.14, 0.95, 0.93, 0.82 and 0.70% per day, respectively. The results of micronucleus experiment and single cell gel electrophoresis showed that cyantraniliprole damaged DNA in liver cells of tilapia larvae. Quantitative PCR results showed that cyantraniliprole could induce the up-regulation of Rpa 3 that is responsible for the DNA repair. The significantly down-regulation of Chk 2 gene was related to p53 pathway. It is therefore proposed that cyantraniliprole causes DNA damage in liver cells of tilapia and activates DNA damage and repair pathways.


Subject(s)
DNA Damage/drug effects , Insecticides/toxicity , Liver/pathology , Pyrazoles/toxicity , Tilapia/embryology , Tilapia/growth & development , ortho-Aminobenzoates/toxicity , Animals , Checkpoint Kinase 2/biosynthesis , DNA Damage/genetics , DNA Repair/drug effects , DNA Repair/genetics , DNA-Binding Proteins/biosynthesis , Gills/metabolism , Hepatocytes/drug effects , Larva , Lethal Dose 50 , Seafood
3.
Sci Rep ; 8(1): 13672, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209405

ABSTRACT

Doxorubicin is one of the most effective chemotherapy drugs used against solid tumors in the treatment of several cancer types. Two different mechanisms, (i) intercalation of doxorubicin into DNA and inhibition of topoisomerase II leading to changes in chromatin structure, (ii) generation of free radicals and oxidative damage to biomolecules, have been proposed to explain the mode of action of this drug in cancer cells. A genome-wide integrative systems biology approach used in the present study to investigate the long-term effect of doxorubicin in Saccharomyces cerevisiae cells indicated the up-regulation of genes involved in response to oxidative stress as well as in Rad53 checkpoint sensing and signaling pathway. Modular analysis of the active sub-network has also revealed the induction of the genes significantly associated with nucleosome assembly/disassembly and DNA repair in response to doxorubicin. Furthermore, an extensive re-wiring of the metabolism was observed. In addition to glycolysis, and sulfate assimilation, several pathways related to ribosome biogenesis/translation, amino acid biosynthesis, nucleotide biosynthesis, de novo IMP biosynthesis and one-carbon metabolism were significantly repressed. Pentose phosphate pathway, MAPK signaling pathway biological processes associated with meiosis and sporulation were found to be induced in response to long-term exposure to doxorubicin in yeast cells.


Subject(s)
DNA, Fungal/drug effects , Doxorubicin/pharmacology , Saccharomyces cerevisiae/metabolism , Topoisomerase II Inhibitors/pharmacology , Transcription, Genetic/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/biosynthesis , Checkpoint Kinase 2/genetics , Chromatin Assembly and Disassembly/drug effects , DNA Repair/genetics , Fermentation/drug effects , Glycolysis/drug effects , Nucleosomes/metabolism , Oxidative Stress/genetics , Pentose Phosphate Pathway/drug effects , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics
4.
Gynecol Oncol ; 150(1): 136-142, 2018 07.
Article in English | MEDLINE | ID: mdl-29804637

ABSTRACT

OBJECTIVE: To analyze the expression and clinical role of CHK1 and CHK2 in metastatic high-grade serous carcinoma (HGSC). METHODS: HGSC effusions (n = 335; 280 peritoneal, 55 pleural) were analyzed for protein expression of total CHK1 and its phosphorylated forms p-ser317 and p-ser296, as well as total CHK2 and its phosphorylated form p-thr68 using immunohistochemistry. Expression was analyzed for association with clinicopathologic parameters, including chemotherapy response, and survival. RESULTS: Carcinoma cells stained positive, predominantly at the nuclei, in the majority of cases (range 83-100% for the five antibodies), while expression in reactive mesothelial cells and tumor-associated macrophages was more variable. Total CHK1 (p = 0.037), p-CHK1ser317 (p = 0.001), p-CHK1ser296 (p = 0.002) and p-CHK2thr68 (p < 0.001) expression was significantly higher in post-chemotherapy disease recurrence compared to pre-chemotherapy effusions obtained at diagnosis. CHK1, p-CHK1ser296, p-CHK2thr68 and p-CHK1ser317 nuclear expression was positively related to expression of the checkpoint regulator WEE1, previously studied in this cohort (p = 0.003, p = 0.013, p = 0.001 and p = 0.01, respectively). Higher total CHK1 (p = 0.007), p-CHK1ser317 (p = 0.004), CHK2 (p = 0.01) and p-CHK2thr68 (p = 0.048) expression was significantly related to shorter overall survival in univariate analysis, and CHK1ser317 was an independent prognostic marker in multivariate analysis (p = 0.025). Higher p-CHK1ser317 (p = 0.03) and CHK2 (p = 0.034) expression was additionally associated with poor progression-free survival. CONCLUSIONS: CHK1 and CHK2 and their activated forms are frequently expressed in HGSC effusions, with higher expression following exposure to chemotherapy, and their expression is related to survival.


Subject(s)
Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/biosynthesis , Checkpoint Kinase 2/metabolism , Cystadenocarcinoma, Serous/enzymology , Adult , Aged , Aged, 80 and over , Checkpoint Kinase 1/biosynthesis , Checkpoint Kinase 1/genetics , Checkpoint Kinase 2/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Enzyme Activation , Female , Humans , Middle Aged , Neoplasm Grading , Survival Analysis , Young Adult
5.
Med Sci Monit ; 23: 4391-4400, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28894083

ABSTRACT

BACKGROUND Previous studies revealed physiological and pathogenetic similarity between vascular smooth muscles cells with severe pulmonary arterial hypertension and tumors. The DNA damage response was found in both pulmonary arterial hypertension (PAH) cells and tumors. The ataxia-telangiectasia mutated proteins (ATM) pathway is considered an important factor in the DNA damage response of tumor formation, but its function in the development of PAH remains unknown. MATERIAL AND METHODS The Sprague-Dawley rat PAH model was established. Three weeks (Group M1), 5 weeks (Group M2), and 7 weeks (Group M3) after drug injection, pulmonary expression of ATM, Checkpoint kinase 2 (Chk2), P53, and P21 were measured. A section of the lungs from Group M2 was used for pulmonary artery vascular smooth muscles cells (PA-SMCs) isolation and culture. The effect of KU60019 in the proliferation and apoptosis of primary cultured rat PA-SMCs was measured by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and TdT-mediated dUTP nick-end labeling (TUNEL), respectively. RESULTS Immunohistochemistry results show that the expression of ATM, Chk2, and P21 increased in Groups M1 and M2, and decreased in Group M3. Additionally, expression of P53 increased in Group M1, and decreased in Groups M2 and M3. RT-PCR and Western blotting demonstrated that in Groups M1 and M2, the expression of ATM, Chk2, P53, and P21 increased, whereas it decreased in Group M3. In cell culture, 0.3 µM and 0.5 µM KU60019 increased the growth of PA-SMCs, and 0.5 µM KU60019 reduced cell apoptosis. CONCLUSIONS Expression of the ATM-Chk2 pathway increased in early stages of PAH formation, but decreased in late stages. In primary cultured PA-SMCs, KU60019 increased cell proliferation and inhibited cell apoptosis.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Mutation , Animals , Apoptosis/drug effects , Ataxia Telangiectasia , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Cell Proliferation/drug effects , Checkpoint Kinase 2/biosynthesis , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Disease Models, Animal , Morpholines/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Thioxanthenes/pharmacology , Tumor Suppressor Protein p53/metabolism
6.
J Mol Biol ; 429(16): 2463-2473, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28712951

ABSTRACT

In a previous study, we identified several novel targets of Dnmt3b using a chromatin library from retinoic acid (RA)-treated P19 cells. The present study describes the regulation of expression and function of checkpoint kinase (Chk2), which was one of the target genes of Dnmt3b. Chromatin immunoprecipitation followed by quantitative PCR analysis showed that recruitment of Dnmt3b on Chk2 promoter is induced following RA treatment of P19 cells. Both bisulfite genomic sequence and COBRA analyses showed that the methylation level of Chk2 promoter is progressively increased during RA-induced neuronal differentiation of P19 cells. Concomitantly, both mRNA and protein expression of Chk2 are reduced as determined by real-time PCR and Western blot analysis, respectively. Suppression of Dnmt3b expression by lentiviral-mediated shRNA resulted in increased expression and reduced methylation of Chk2, which clearly showed that Dnmt3b is responsible for transcriptional silencing of Chk2 gene in RA-treated P19 cells. Neuronal differentiation of P19 cells was inhibited upon enforced Chk2 expression in P19 cells, which showed that the decrease in endogenous expression of Chk2 is essential for normal differentiation. Ectopic Chk2 expression also negatively regulated cell cycle arrest and apoptosis following RA treatment, which could also contribute to impaired neuronal differentiation. Together, this study described the regulation of Chk2 expression through promoter methylation and also presented a novel role of Chk2 during neuronal differentiation, which is independent of its previously known function in DNA damage response.


Subject(s)
Cell Differentiation , Checkpoint Kinase 2/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Expression Regulation , Neurons/physiology , Promoter Regions, Genetic , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Humans , Mice , Neurons/drug effects , Real-Time Polymerase Chain Reaction , Tretinoin/metabolism , DNA Methyltransferase 3B
7.
Int J Mol Sci ; 17(11)2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27801830

ABSTRACT

Checkpoint kinase 2 (CHK2) and cell division cycle 25C (CDC25C) are two proteins involved in the DNA damage response pathway, playing essential roles in maintaining genome integrity. As one of the major hallmarks of abnormal cellular division, genomic instability occurs in most cancers. In this study, we identified the functional expression of pCHK2-Thr68 and pCDC25C-Ser216 in breast cancer, as well as its association with breast cancer survival. Tissue microarray analysis using immunohistochemistry was constructed to identify the expression of pCHK2-Thr68 and pCDC25C-Ser216 in 292 female breast cancer patients. The relationship among protein expression, clinicopathological factors (e.g., human epidermal growth factor receptor 2 (HER 2), tumor size, tumor-node-metastasis (TNM) classification), and overall survival of the breast cancer tissues were analyzed using Pearson's χ-square (χ²) test, Fisher's exact test, multivariate logistic regression and Kaplan-Meier survival analysis. Significantly higher expressions of pCHK2-Thr68 and pCDC25C-Ser216 were observed in the nucleus of the breast cancer cells compared to the paracancerous tissue (pCHK2-Thr68, 20.38% vs. 0%; pCDC25C-Ser216, 82.26% vs. 24.24%). The expression of pCHK2-Thr68 and pCDC25C-Ser216 in breast cancer showed a positive linear correlation (p = 0.026). High expression of pCHK2-Thr68 was associated with decreased patient survival (p = 0.001), but was not an independent prognostic factor. Our results suggest that pCHK2-Thr68 and pCDC25C-Ser216 play important roles in breast cancer and may be potential treatment targets.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Checkpoint Kinase 2/biosynthesis , cdc25 Phosphatases/biosynthesis , Adult , Aged , Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Checkpoint Kinase 2/genetics , DNA Damage/genetics , Female , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Microarray Analysis , Middle Aged , Neoplasm Staging , Prognosis , Receptor, ErbB-2/genetics , cdc25 Phosphatases/genetics
8.
Oncol Rep ; 33(6): 2999-3005, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25962492

ABSTRACT

The present study aimed to investigate the influence of SHP-1 on the radioresistance of the nasopharyngeal carcinoma (NPC) cell line CNE-2 and the relevant underlying mechanisms. The human NPC cell line CNE-2 was transfected with a lentivirus that contained the SHP-1 gene or a nonsense sequence (referred to as LP-H1802Lv201 and LP-NegLv201 cells, respectively). Cells were irradiated with different ionizing radiation (IR) doses. Cell survival, DNA double-strand breaks (DSBs), apoptosis, cell cycle distribution, and the expression of related proteins were assessed using colony formation assay, immunofluorescent assays (IFAs), flow cytometry (FCM) and western blot analyses, respectively. Compared with the control (CNE-2 cells) and LP-NegLv201 cells, LP-H1802Lv201 cells were more resistant to IR. IFAs showed that IR caused less histone H2AX phosphorylation (γH2AX) and RAD51 foci in the LP-H1802Lv201 cells. Compared with the control and LP-NegLv201 cells, LP-H1802Lv201 cells showed increased S phase arrest. After IR, the apoptotic rate of the LP-H1802Lv201 cells was lower in contrast to the control and LP-NegLv201 cells. Western blot analyses showed that IR increased the phosphorylation of ataxia telangiectasia mutated (ATM) kinase, checkpoint kinase 2 (CHK2), ataxia telangiectasia and Rad3-related (ATR) protein, checkpoint kinase 1 (CHK1) and p53. In LP-H1802Lv201 cells, the phosphorylation levels of ATM and CHK2 were significantly increased while the p53 phosphorylation level was decreased compared to these levels in the control and LP-NegLv201 cells. Phosphorylation of ATR and CHK1 did not show significant differences in the three cell groups. Overexpression of SHP-1 in the CNE-2 cells led to radioresistance and the radioresistance was related to enhanced DNA DSB repair, increased S phase arrest and decreased cell apoptosis.


Subject(s)
Checkpoint Kinase 2/metabolism , Nasopharyngeal Neoplasms/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Radiation Tolerance/genetics , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinoma , Cell Cycle Checkpoints/radiation effects , Checkpoint Kinase 2/biosynthesis , DNA Breaks, Double-Stranded/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/radiotherapy , Phosphorylation/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/biosynthesis , Radiation, Ionizing
9.
Tumour Biol ; 36(8): 6095-101, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25773391

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs of 19-25 nt that can regulate gene expression at a posttranscriptional level. Increasing evidence indicates that miRNAs participate in almost every step of cellular processes and are often aberrantly expressed in human cancer. The aim of this study was to investigate the functional significance of miR-191 and to identify its possible target genes in osteosarcoma cells. Here, we found that the expression level of miR-191 was increased in osteosarcoma tissues in comparison with the adjacent normal tissues. The enforced expression of miR-191 was able to promote cell proliferation in Saos-2 and MG62 cells, while miR-191 antisense oligonucleotides blocked cell proliferation. At the molecular level, our results further revealed that expression of tumor suppressor gene, checkpoint kinase 2, was negatively regulated by miR-191. Therefore, we consider that miR-191 act as an onco-MicroRNA for osteosarcoma and it would offer a new way in molecular targeting cancer treatment.


Subject(s)
Cell Proliferation/genetics , Checkpoint Kinase 2/biosynthesis , MicroRNAs/biosynthesis , Osteosarcoma/genetics , Cell Line, Tumor , Checkpoint Kinase 2/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotides, Antisense/genetics , Osteosarcoma/pathology
10.
Sci Rep ; 5: 7886, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25601385

ABSTRACT

The checkpoint kinase Rad53 is crucial to regulate DNA replication in the presence of replicative stress. Under conditions that interfere with the progression of replication forks, Rad53 prevents Exo1-dependent fork degradation. However, although EXO1 deletion avoids fork degradation in rad53 mutants, it does not suppress their sensitivity to the ribonucleotide reductase (RNR) inhibitor hydroxyurea (HU). In this case, the inability to restart stalled forks is likely to account for the lethality of rad53 mutant cells after replication blocks. Here we show that Rad53 regulates replication restart through the checkpoint-dependent transcriptional response, and more specifically, through RNR induction. Thus, in addition to preventing fork degradation, Rad53 prevents cell death in the presence of HU by regulating RNR-expression and localization. When RNR is induced in the absence of Exo1 and RNR negative regulators, cell viability of rad53 mutants treated with HU is increased and the ability of replication forks to restart after replicative stress is restored.


Subject(s)
Cell Cycle Proteins/biosynthesis , Checkpoint Kinase 2/biosynthesis , DNA Replication/genetics , Exodeoxyribonucleases/genetics , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Survival/drug effects , Checkpoint Kinase 2/genetics , DNA Repair/genetics , DNA Replication/drug effects , Exodeoxyribonucleases/metabolism , Gene Expression Regulation, Fungal/drug effects , Hydroxyurea/pharmacology , Mutation , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics
11.
Int J Oncol ; 46(2): 597-606, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25405317

ABSTRACT

The mammalian timeless (TIM) protein interacts with proteins of the endogenous clock and essentially contributes to the circadian rhythm. In addition, TIM is involved in maintenance of chromosome integrity, growth control and development. Thus, we hypothesized that TIM may exert a potential protumorigenic function in human hepatocarcinogenesis. TIM was overexpressed in a subset of human HCCs both at the mRNA and the protein level. siRNA-mediated knockdown of TIM reduced cell viability due to the induction of apoptosis and G2 arrest. The latter was mediated via CHEK2 phosphorylation. In addition, siRNA-treated cells showed a significantly reduced migratory capacity and reduced expression levels of various proteins. Mechanistically, TIM directly interacts with the eukaryotic elongation factor 1A2 (EEF1A2), which binds to actin filaments to promote tumor cell migration. siRNA-mediated knockdown of TIM reduced EEF1A2 protein levels thereby affecting ribosomal protein biosynthesis. Thus, overexpression of TIM exerts oncogenic function in human HCCs, which is mediated via CHEK2 and EEF1A2.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Adolescent , Adult , Aged , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/biosynthesis , Checkpoint Kinase 2/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/biosynthesis , Liver Neoplasms/pathology , Male , Middle Aged , Peptide Elongation Factor 1/biosynthesis , RNA, Small Interfering
12.
BMC Cancer ; 14: 287, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24758542

ABSTRACT

BACKGROUND: The DNA damage checkpoint signalling cascade sense damaged DNA and coordinates cell cycle arrest, DNA repair, and/or apoptosis. However, it is still not well understood how the signalling system differentiates between different kinds of DNA damage. N-nitroso-N-ethylurea (NEU), a DNA ethylating agent induces both transversions and transition mutations. METHODS: Immunoblot and comet assays were performed to detect DNA breaks and activation of the canonical checkpoint signalling kinases following NEU damage upto 2 hours. To investigate whether mismatch repair played a role in checkpoint activation, knock-down studies were performed while flow cytometry analysis was done to understand whether the activation of the checkpoint kinases was cell cycle phase specific. Finally, breast epithelial cells were grown as 3-dimensional spheroid cultures to study whether NEU can induce upregulation of vimentin as well as disrupt cell polarity of the breast acini, thus causing transformation of epithelial cells in culture. RESULTS: We report a novel finding that NEU causes activation of major checkpoint signalling kinases, Chk1 and Chk2. This activation is temporally controlled with Chk2 activation preceding Chk1 phosphorylation, and absence of cross talk between the two parallel signalling pathways, ATM and ATR. Damage caused by NEU leads to the temporal formation of both double strand and single strand breaks. Activation of checkpoints following NEU damage is cell cycle phase dependent wherein Chk2 is primarily activated during G2-M phase whilst in S phase, there is immediate Chk1 phosphorylation and delayed Chk2 response. Surprisingly, the mismatch repair system does not play a role in checkpoint activation, at doses and duration of NEU used in the experiments. Interestingly, NEU caused disruption of the well-formed polarised spheroid archithecture and upregulation of vimentin in three-dimensional breast acini cultures of non-malignant breast epithelial cells upon NEU treatment indicating NEU to have the potential to cause early transformation in the cells. CONCLUSION: NEU causes damage in mammalian cells in the form of double strand and single strand breaks that temporally activate the major checkpoint signalling kinases without the occurrence of cross-talk between the pathways. NEU also appear to cause transformation in three-dimensional spheroid cultures.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Checkpoint Kinase 2/biosynthesis , Protein Kinases/biosynthesis , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle Checkpoints/drug effects , Cell Transformation, Neoplastic/genetics , Checkpoint Kinase 1 , Checkpoint Kinase 2/metabolism , DNA Damage/drug effects , DNA Repair/drug effects , Ethylnitrosourea/pharmacology , Humans , Phosphorylation , Protein Kinases/metabolism , Signal Transduction/drug effects
13.
Pathobiology ; 81(1): 25-35, 2014.
Article in English | MEDLINE | ID: mdl-23969480

ABSTRACT

OBJECTIVES: The aims of this study were to assess expressions of the DNA damage response (DDR)-related proteins and to investigate their clinical significances in gastric carcinoma. METHODS: Two independent cohorts, a training set (n=524) and validation set (n=394), of gastric cancer patients were enrolled. Ataxia telangiectasia mutated (ATM), checkpoint kinase 2 (Chk2), and p53 expressions were examined by immunohistochemistry using tissue microarray. RESULTS: ATM loss, Chk2 loss, and p53 positivity were observed in 21.8, 14.1, and 36.1% of the training set, and in 17.3, 12.2, and 35.8% of the validation set, respectively. In the training set, the aberrant expressions of ATM, Chk2, or p53 were significantly associated with an advanced TNM stage and poor disease-specific survival. This association was verified in the validation set. Chk2 positivity and p53 negativity were significantly related to a prolonged disease-specific survival. Also, patients with nonaberrant expressional levels of all 3 DDR-related proteins had a more favorable outcome than others. Multivariate analyses showed that Chk2 loss and at least 1 aberrant DDR-related protein remained as independent prognostic factors of poor disease-specific survival. CONCLUSIONS: This study elucidated the prognostic implications of DDR-related proteins, and suggests that their aberrant expressions play critical roles in the development and progression of gastric cancer.


Subject(s)
Adenocarcinoma/metabolism , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Checkpoint Kinase 2/biosynthesis , DNA Damage , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Ataxia Telangiectasia Mutated Proteins/analysis , Biomarkers, Tumor/analysis , Checkpoint Kinase 2/analysis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis , Tumor Suppressor Protein p53/analysis
14.
Biochem Biophys Res Commun ; 441(3): 593-9, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24183725

ABSTRACT

Meiotic cells generate physiological programmed DNA double-strand breaks (DSBs) to initiate meiotic recombination. Interhomolog repair of the programmed DSBs by meiotic recombination is vital to ensure accurate chromosome segregation at meiosis I to produce normal gametes. In budding yeast, the DNA damage checkpoint kinase Rad53 is activated by DSBs which accidentally occur as DNA lesions in mitosis and meiosis; however, meiotic programmed DSBs which occur at approximately 160 loci per genome fail to activate the kinase. Thus, Rad53 activation appears to be silenced in response to meiotic programmed DSBs. In this study, to address the biological significance of Rad53's insensitivity to meiotic DSBs, we examined the effects of Rad53 overexpression on meiotic processes. The overexpression led to partial activation of Rad53, uncovering that the negative impacts of Rad53 kinase activation on meiotic progression, and formation and interhomolog repair of meiotic programmed DSBs.


Subject(s)
Cell Cycle Proteins/biosynthesis , Checkpoint Kinase 2/biosynthesis , DNA Breaks, Double-Stranded , DNA Repair , Recombination, Genetic , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae/enzymology , Cell Cycle Proteins/genetics , Checkpoint Kinase 2/genetics , Meiosis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
15.
Oncol Rep ; 30(5): 2467-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002546

ABSTRACT

The aim of the present study was to evaluate the antitumor mechanism of dihydromyricetin (DHM). Results showed that DHM significantly inhibited cell viability of HepG2 and Hep3B cells in a dose-dependent manner. DHM induced G2/M cell-cycle arrest in HepG2 and Hep3B cells by altering the expression of cell cycle proteins such as cyclin A, cyclin B1, Cdk1, p53, Cdc25c, p-Cdc25c Chk1 and Chk, which are critical for G2/M transition. Knockdown of p53 and Chk1 in HepG2 cells did not affect G2/M phase arrest caused by DHM. Furthermore, G2/M arrest induced by DHM can be disrupted by Chk2 siRNA. These findings indicate that DHM inhibits the growth of hepatocellular carcinoma (HCC) cells via G2/M phase cell cycle arrest through Chk1/Chk2/Cdc25C pathway. The present study identified effects of DHM in G2/M phase arrest in HCC and described detailed mechanisms of G2/M phase arrest by this agent, which may contribute to its overall cancer preventive efficacy in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Checkpoint Kinase 2/genetics , Liver Neoplasms/drug therapy , Protein Kinases/genetics , cdc25 Phosphatases/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2/biosynthesis , Flavonols/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Protein Kinases/biosynthesis , Signal Transduction/drug effects , cdc25 Phosphatases/biosynthesis
16.
Mol Cancer Ther ; 12(10): 2078-87, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939379

ABSTRACT

Ataxia-telangiectasia mutated (ATM) is a major regulator of the DNA damage response. ATM promotes the activation of BRCA1, CHK2, and p53 leading to the induction of response genes such as CDKN1A (p21), GADD45A, and RRM2B that promote cell-cycle arrest and DNA repair. The upregulation of these response genes may contribute to resistance of cancer cells to genotoxic therapies. Here, we show that histone deacetylases (HDAC) play a major role in mitigating the response of the ATM pathway to DNA damage. HDAC inhibition decreased ATM activation and expression, and attenuated the activation of p53 in vitro and in vivo. Select depletion of HDAC1 and HDAC2 was sufficient to modulate ATM activation, reduce GADD45A and RRM2B induction, and increase sensitivity to DNA strand breaks. The regulation of ATM by HDAC enzymes therefore suggests a vital role for HDAC1 and HDAC2 in the DNA damage response, and the potential use of the ATM pathway as a pharmacodynamic marker for combination therapies involving HDAC inhibitors.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Ataxia Telangiectasia Mutated Proteins/biosynthesis , BRCA1 Protein/biosynthesis , Checkpoint Kinase 2/biosynthesis , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Humans , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Tumor Suppressor Protein p53/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...