Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Histochem Cytochem ; 70(3): 199-210, 2022 03.
Article in English | MEDLINE | ID: mdl-34978208

ABSTRACT

Current scientific literature lacks data on the prognostic value of the expression of RAD51 and BRCA2 in gastric adenocarcinoma. Therefore, we aimed to evaluate those and other homologous recombination-related proteins (ATM, ATR, BRCA1, CHK2, γH2AX, p53) in gastric cancer, assessing their correlation with clinical prognosis. Paraffin-embedded samples were obtained from surgical specimens collected in total or subtotal gastrectomy procedures. Between 2008 and 2017, 121 patients with advanced gastric adenocarcinoma underwent surgical resection and were included in this study. Negativity for nuclear RAD51 correlated with vascular invasion, lymph node metastasis, larger tumor size, and lower overall survival and disease-free survival in univariate analysis. However, nuclear RAD51-negative cases presented better response rates to adjuvant therapy than the positive ones. Nuclear ATR negativity correlated with larger tumor size and a higher histological grade. Positivity for ATM was associated with more prolonged disease-free survival. Positivity for nuclear BRCA2 correlated with lower overall survival and diffuse histological type, whereas its high expression was associated with vascular invasion. Nevertheless, tumors positive for nuclear BRCA2 were more frequently low grade in the intestinal histological type. Our findings indicate that RAD51 and BRCA2 are valuable immunohistochemical prognostic markers in gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/diagnosis , BRCA2 Protein/analysis , Rad51 Recombinase/analysis , Stomach Neoplasms/diagnosis , Adenocarcinoma/metabolism , BRCA2 Protein/biosynthesis , Cohort Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Rad51 Recombinase/biosynthesis , Retrospective Studies , Stomach Neoplasms/metabolism
2.
Appl Immunohistochem Mol Morphol ; 29(4): 270-276, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33417321

ABSTRACT

Breast cancer is a heterogeneous disease at morphologic and molecular levels, which is considered the most commonly occurring cancer in women. RAD51, a DNA-repairing protein, involves homologous recombination and has a vital role in genome stability. Polymorphism of the RAD51 gene, and its overexpression, has been proposed to be associated with the development of breast cancer. Overexpression of RAD51 in many types of human cancer including metastatic breast cancer may signify its potential use as a biomarker. Considering the numerous reports on the role of the 5'-UTR-RAD51 polymorphism in breast cancer, this study aimed to investigate the utility of RAD51 gene expression and its variants G135C and G172T as a possible foretelling factor of breast cancer development. DNA sequencing and immunohistochemistry of RAD51 were conducted on 103 samples from patients diagnosed with sporadic breast cancer and 80 samples from a control group. The results demonstrated that the RAD51 variants, G135C and G172T, were significantly presented in the breast cancer tissue compared with the control group. RAD51 expression was mainly shown in the cytoplasm of malignant cells (56% of cases) and significantly correlated with p53 and G135C, C135C variants. Moreover, the occurrence of the G172T variant was significantly associated with the expression of estrogen receptor. Interestingly, 21/26 (81%) of the triple-negative breast cancer showed G135C and C135C genotypes that were significantly associated with the expression of RAD51 (73%). In conclusion, the G135C and C135C variants together with the cytoplasmic expression of RAD51 may have clinical potential as a prognostic predictor for breast cancer development and aggressiveness.


Subject(s)
5' Untranslated Regions , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins , Polymorphism, Single Nucleotide , Rad51 Recombinase , Triple Negative Breast Neoplasms , Adult , Case-Control Studies , Female , Humans , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
J Cell Physiol ; 236(7): 4944-4953, 2021 07.
Article in English | MEDLINE | ID: mdl-33368268

ABSTRACT

Obesity causes many reproductive dysfunctions such as reduced conception, infertility, and early pregnancy loss, and this is largely due to the negative effects of obesity on oocyte and embryo quality. In the present study, we employed single-cell RNA transcriptome sequencing to investigate the potential causes for the maternal obesity effects on mouse embryos. Our results showed that the 4-cell and morula/blastocyst rates were all significantly decreased during embryo development in obese mice. Genome-wide analysis indicated that obesity altered the expression of more than 1100 genes in 2-cell embryos, including the genes which were related to the p53 signaling pathway and apoptosis. Further analysis showed that the expression of 47 genes related to DNA damage was changed, and a positive γH2A signal and the altered expression of Rad51 and Tex15 were observed in the obese embryos. Obesity also affected histone methylation, shown by the decrease of the H3K4-me2 level. Besides this, we observed the occurrence of autophagy and apoptosis in the embryos of obese mice. There were 42 genes that were related to autophagy/apoptosis that showed aberrant expression, and the positive LC3 signal and the decrease of Clec16a, Rraga, and Atg10 level were also observed. In summary, our study suggested that obesity affected early embryonic development by inducing DNA damage, aberrant histone methylation, and autophagy levels in mice.


Subject(s)
Autophagy/physiology , DNA Methylation/genetics , DNA Repair/genetics , Embryonic Development/physiology , Obesity, Maternal/pathology , Animals , Apoptosis/physiology , Blastocyst/physiology , Cell Cycle Proteins/biosynthesis , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Oocytes/cytology , Pregnancy , Rad51 Recombinase/biosynthesis , Single-Cell Analysis , Transcriptome
4.
Cell Rep ; 33(12): 108543, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33357432

ABSTRACT

DNA damage tolerance (DDT) and homologous recombination (HR) stabilize replication forks (RFs). RAD18/UBC13/three prime repair exonuclease 2 (TREX2)-mediated proliferating cell nuclear antigen (PCNA) ubiquitination is central to DDT, an error-prone lesion bypass pathway. RAD51 is the recombinase for HR. The RAD51 K133A mutation increased spontaneous mutations and stress-induced RF stalls and nascent strand degradation. Here, we report in RAD51K133A cells that this phenotype is reduced by expressing a TREX2 H188A mutation that deletes its exonuclease activity. In RAD51K133A cells, knocking out RAD18 or overexpressing PCNA reduces spontaneous mutations, while expressing ubiquitination-incompetent PCNAK164R increases mutations, indicating DDT as causal. Deleting TREX2 in cells deficient for the RF maintenance proteins poly(ADP-ribose) polymerase 1 (PARP1) or FANCB increased nascent strand degradation that was rescued by TREX2H188A, implying that TREX2 prohibits degradation independent of catalytic activity. A possible explanation for this occurrence is that TREX2H188A associates with UBC13 and ubiquitinates PCNA, suggesting a dual role for TREX2 in RF maintenance.


Subject(s)
DNA Replication , Exodeoxyribonucleases/metabolism , Mutation , Phosphoproteins/metabolism , Rad51 Recombinase/metabolism , Animals , Exodeoxyribonucleases/genetics , Humans , Male , Mice , Phosphoproteins/genetics , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Transfection
5.
Cell Transplant ; 29: 963689720931433, 2020.
Article in English | MEDLINE | ID: mdl-32830515

ABSTRACT

Genetic instability, raised from dysregulation of DNA repair, is involved in tumor development. OTUB2 (ovarian tumor domain protease domain-containing ubiquitin aldehyde-binding protein 2), which is responsible for DNA double-strand break (DSB), is implicated in carcinogenesis of various tumors. The effect of OTUB2 on endometrial cancer progression was then investigated. First, OTUB2 was found to be upregulated in endometrial cancer tissues and cell lines, and was closely associated with overall survival of endometrial cancer patients. Cell Counting Kit-8 and flow cytometry assay results revealed that overexpression of OTUB2 enhanced cell viability of endometrial cancer cells, while knockdown of OTUB2 inhibited cell viability. Moreover, as demonstrated by promoting cell viability and suppression of cell apoptosis, cisplatin-induced cell damage was reversed by OTUB2. Mechanistically, OTUB2 could activate Yes-associated protein/transcriptional co-activator with PDZ-binding motif (TAZ) to promote homologous recombination repair via depletion of γH2AX (phosphorylation of histone H2AX) and accumulation of Rad51. In vivo xenograft model also showed that silence of OTUB2 suppressed the growth of endometrial cancer and increased tumor sensitivity to antitumor drugs. In conclusion, OTUB2 promoted homologous recombination repair in endometrial cancer via YAP/TAZ-mediated Rad51 expression, providing a potential therapeutic target for endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Rad51 Recombinase/biosynthesis , Recombinational DNA Repair , Thiolester Hydrolases/metabolism , Animals , Cell Line, Tumor , Disease Progression , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Thiolester Hydrolases/genetics , Transfection
6.
BMB Rep ; 52(2): 151-156, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30638176

ABSTRACT

RAD51 recombinase plays a critical role in homologous recombination and DNA damage repair. Here we showed that expression of RAD51 is frequently upregulated in lung cancer tumors compared with normal tissues and is associated with poor survival (hazard ratio (HR) = 2, P = 0.0009). Systematic investigation of lung cancer cell lines revealed higher expression of RAD51 in KRAS mutant (MT) cells compared to wildtype (WT) cells. We further showed that MT KRAS, but not WT KRAS, played a critical role in RAD51 overexpression via MYC. Moreover, our results revealed that KRAS MT cells are highly dependent on RAD51 for survival and depletion of RAD51 resulted in enhanced DNA double strand breaks, defective colony formation and cell death. Together, our results suggest that mutant KRAS promotes RAD51 expression to enhance DNA damage repair and lung cancer cell survival, suggesting that RAD51 may be an effective therapeutic target to overcome chemo/radioresistance in KRAS mutant cancers. [BMB Reports 2019; 52(2): 151-156].


Subject(s)
DNA Damage , DNA Repair , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Rad51 Recombinase/biosynthesis , Cell Line, Tumor , Cell Survival/physiology , DNA Breaks, Double-Stranded , Homologous Recombination , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Rad51 Recombinase/genetics , Up-Regulation
7.
Curr Cancer Drug Targets ; 19(6): 479-494, 2019.
Article in English | MEDLINE | ID: mdl-30182856

ABSTRACT

BACKGROUND: c-Myc plays a major role in the maintenance of glycolytic metabolism and hematopoietic stem cell (HSC) quiescence. OBJECTIVE: Targeting modulators of HSC quiescence and metabolism could lead to HSC cell cycle entry with concomitant expansion. METHODS AND RESULTS: Here we show that c-Myc inhibitor 10074-G5 treatment leads to 2-fold increase in murine LSKCD34low HSC compartment post 7 days. In addition, c-Myc inhibition increases CD34+ and CD133+ human HSC number. c-Myc inhibition leads to downregulation of glycolytic and cyclindependent kinase inhibitor (CDKI) gene expression ex vivo and in vivo. In addition, c-Myc inhibition upregulates major HDR modulator Rad51 expression in hematopoietic cells. Besides, c-Myc inhibition does not alter proliferation kinetics of endothelial cells, fibroblasts or adipose-derived mesenchymal stem cells, however, it limits bone marrow derived mesenchymal stem cell proliferation. We further demonstrate that a cocktail of c-Myc inhibitor 10074-G5 along with tauroursodeoxycholic acid (TUDCA) and i-NOS inhibitor L-NIL provides a robust HSC maintenance and expansion ex vivo as evident by induction of all stem cell antigens analyzed. Intriguingly, the cocktail of c-Myc inhibitor 10074-G5, TUDCA and L-NIL improves HDR related gene expression. CONCLUSION: These findings provide tools to improve ex vivo HSC maintenance and expansion, autologous HSC transplantation and gene editing through modulation of HSC glycolytic and HDR pathways.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Oxadiazoles/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Rad51 Recombinase/metabolism , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Cell Culture Techniques , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Lysine/analogs & derivatives , Lysine/pharmacology , Mice , Mice, Inbred BALB C , Mice, SCID , Nitric Oxide Synthase/antagonists & inhibitors , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Small Molecule Libraries/pharmacology , Taurochenodeoxycholic Acid/pharmacology
8.
Nucleic Acids Res ; 46(17): 8898-8907, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30032296

ABSTRACT

BRCA proteins are essential for homologous recombination (HR) DNA repair, and their germline or somatic inactivation is frequently observed in human tumors. Understanding the molecular mechanisms underlying the response of BRCA-deficient tumors to chemotherapy is paramount for developing improved personalized cancer therapies. While PARP inhibitors have been recently approved for treatment of BRCA-mutant breast and ovarian cancers, not all patients respond to this therapy, and resistance to these novel drugs remains a major clinical problem. Several mechanisms of chemoresistance in BRCA2-deficient cells have been identified. Rather than restoring normal recombination, these mechanisms result in stabilization of stalled replication forks, which can be subjected to degradation in BRCA2-mutated cells. Here, we show that the transcriptional repressor E2F7 modulates the chemosensitivity of BRCA2-deficient cells. We found that BRCA2-deficient cells are less sensitive to PARP inhibitor and cisplatin treatment after E2F7 depletion. Moreover, we show that the mechanism underlying this activity involves increased expression of RAD51, a target for E2F7-mediated transcriptional repression, which enhances both HR DNA repair, and replication fork stability in BRCA2-deficient cells. Our work describes a new mechanism of therapy resistance in BRCA2-deficient cells, and identifies E2F7 as a putative biomarker for tumor response to PARP inhibitor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA2 Protein/deficiency , Drug Resistance, Neoplasm/physiology , E2F7 Transcription Factor/physiology , Enzyme Inhibitors/pharmacology , Neoplasm Proteins/physiology , CRISPR-Cas Systems , Cell Line, Tumor , DNA Replication/drug effects , DNA Replication/physiology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , E2F7 Transcription Factor/deficiency , Gene Knockout Techniques , Genes, BRCA2 , Humans , Neoplasm Proteins/deficiency , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerases , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/physiology
9.
J Cancer Res Clin Oncol ; 144(7): 1329-1337, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29777301

ABSTRACT

PURPOSE: To determine if inhibiting neuropilin-1 (NRP-1) affects the radiosensitivity of NSCLC cells through a vascular endothelial growth factor receptor 2 (VEGFR2)-independent pathway, and to assess the underlying mechanisms. METHODS: The expression of VEGFR2, NRP-1, related signaling molecules, abelson murine leukemia viral oncogene homolog 1 (ABL-1), and RAD51 were determined by RT-PCR and Western blotting, respectively. Radiosensitivity was assessed using the colony-forming assay, and the cell apoptosis were analyzed by flow cytometry. RESULTS: We selected two cell lines with high expression levels of VEGFR2, including Calu-1 cells that have high NRP-1 expression, and H358 cells that have low NRP-1 expression. Upon inhibition of p-VEGFR2 by apatinib in Calu-1 cells, the expression of NRP-1 protein and other related proteins in the pathway was still high. Upon NRP-1 siRNA treatment, the expression of both NRP-1 and RAD51 decreased (p < 0.01; p < 0.05). Upon ABL-1 siRNA treatment, the expression of NRP-1 was increased and the expression of RAD51 was unchanged. Calu-1 cells treated with NRP-1 siRNA exhibited significantly higher apoptosis and radiation sensitivity in radiation therapy compared to Calu-1 cells treated with apatinib alone (p < 0.01; p < 0.01). The apoptosis and radiation sensitivity in H358 cells with NRP-1 overexpression was similar to the control group regardless of VEGFR2 inhibition. CONCLUSIONS: We demonstrated that when VEGFR2 was inhibited, NRP-1 appeared to regulate RAD51 expression through the VEGFR2-independent ABL-1 pathway, consequently regulating radiation sensitivity. In addition, the combined inhibition of VEGFR2 and NRP-1 appears to sensitize cancer cells to radiation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/biosynthesis , Pyridines/pharmacology , Rad51 Recombinase/biosynthesis , Radiation Tolerance/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
10.
Oncology ; 95(1): 20-30, 2018.
Article in English | MEDLINE | ID: mdl-29694959

ABSTRACT

OBJECTIVE: Multimodal treatments that include preoperative platinum-based chemotherapy are fundamental to the treatment of advanced non-small cell lung cancer (NSCLC). This study aimed to investigate the predictive value of DNA repair protein expression in surgically resected NSCLCs in terms of prognosis and responses to platinum-containing chemotherapy. METHODS: This retrospective study included 136 patients with NSCLC who were treated with preoperative platinum-based chemotherapy, followed by curative lung resection. ATM, RAD51, LKB1, H2AX, and SIRT1 expression levels were analyzed in resected tumor specimens via immunostaining and were used to classify patients and compare survival and responses to chemotherapy. RESULTS: SIRT1 expression correlated significantly with improved responses to platinum-based chemotherapy (odds ratio, 2.28; p = 0.024), progression-free survival (hazard ratio [HR], 0.74; p = 0.036), overall survival (HR, 0.63; p = 0.006), and tumor-bearing survival (HR, 0.62; p = 0.014). After adjusting for clinical variables, the HR of SIRT1 expression remained significant for overall survival (HR, 0.59; p = 0.039) but not for progression-free survival (HR, 0.74; p = 0.183). No prognostic stratification was observed for the other 4 markers. CONCLUSION: Patients with SIRT1-expressing NSCLC had superior responses to chemotherapy and longer survival durations than those with SIRT1-negative cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carboplatin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/therapeutic use , Lung Neoplasms/drug therapy , Sirtuin 1/biosynthesis , Sirtuin 1/genetics , AMP-Activated Protein Kinase Kinases , Adult , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Ataxia Telangiectasia Mutated Proteins/genetics , Carboplatin/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Cisplatin/adverse effects , Combined Modality Therapy/methods , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/genetics , Disease-Free Survival , Female , Histones/biosynthesis , Histones/genetics , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Retrospective Studies , Treatment Outcome
11.
Mol Cells ; 40(2): 143-150, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28190324

ABSTRACT

Homologous recombination (HR) is necessary for maintenance of genomic integrity and prevention of various mutations in tumor suppressor genes and proto-oncogenes. Rad51 and Rad54 are key HR factors that cope with replication stress and DNA breaks in eukaryotes. Rad51 binds to single-stranded DNA (ssDNA) to form the presynaptic filament that promotes a homology search and DNA strand exchange, and Rad54 stimulates the strand-pairing function of Rad51. Here, we studied the molecular dynamics of Rad51 and Rad54 during the cell cycle of HeLa cells. These cells constitutively express Rad51 and Rad54 throughout the entire cell cycle, and the formation of foci immediately increased in response to various types of DNA damage and replication stress, except for caffeine, which suppressed the Rad51-dependent HR pathway. Depletion of Rad51 caused severe defects in response to postreplicative stress. Accordingly, HeLa cells were arrested at the G2-M transition although a small amount of Rad51 was steadily maintained in HeLa cells. Our results suggest that cell cycle progression and proliferation of HeLa cells can be tightly controlled by the abundance of HR proteins, which are essential for the rapid response to postreplicative stress and DNA damage stress.


Subject(s)
DNA Damage , DNA Helicases/genetics , Nuclear Proteins/genetics , Rad51 Recombinase/genetics , Cell Cycle/genetics , Cell Division/genetics , DNA Helicases/biosynthesis , DNA-Binding Proteins , G2 Phase/genetics , HeLa Cells , Homologous Recombination , Humans , Nuclear Proteins/biosynthesis , Rad51 Recombinase/biosynthesis , Stress, Physiological/genetics
12.
PLoS Genet ; 12(8): e1006208, 2016 08.
Article in English | MEDLINE | ID: mdl-27513445

ABSTRACT

The RAD51 protein plays a key role in the homology-directed repair of DNA double-strand breaks and is important for maintaining genome stability. Here we report on a novel human RAD51 variant found in an aggressive and therapy-refractive breast carcinoma. Expression of the RAD51 G151D variant in human breast epithelial cells increases the levels of homology-directed repair. Expression of RAD51 G151D in cells also promotes high levels of chromosomal aberrations and sister chromatid exchanges. In vitro, the purified RAD51 G151D protein directly and significantly enhances DNA strand exchange activity in the presence of RPA. In concordance with this result, co-incubation of G151D with BRCA2 resulted in a much higher level of strand-exchange activity compared to WT RAD51. Strikingly, the RAD51 G151D variant confers resistance to multiple DNA damaging agents, including ionizing radiation, mitomycin C, and doxorubicin. Our findings demonstrate that the RAD51 G151D somatic variant has a novel hyper-recombination phenotype and suggest that this property of the protein is important for the repair of DNA damage, leading to drug resistance.


Subject(s)
BRCA2 Protein/genetics , Breast Neoplasms/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , BRCA2 Protein/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Chromosome Aberrations/drug effects , Chromosome Aberrations/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/genetics , Doxorubicin/administration & dosage , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genomic Instability/drug effects , Genomic Instability/radiation effects , Humans , MCF-7 Cells , Mitomycin/administration & dosage , Mutation , Rad51 Recombinase/biosynthesis , Radiation, Ionizing , Sister Chromatid Exchange/genetics
13.
Genes Cells ; 21(7): 789-97, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27251002

ABSTRACT

H2AX is expressed at very low levels in quiescent normal cells in vivo and in vitro. Such cells repair DNA double-strand breaks (DSBs) induced by γ-irradiation through a transient stabilization of H2AX. However, the resultant cells accumulate small numbers of irreparable (or persistent) DSBs via an unknown mechanism. We found that quiescent cells that had repaired DSBs directly induced by γ-rays were prone to accumulate DSBs during the subsequent DNA replication. Unlike directly induced DSBs, secondary DSBs were not efficiently repaired, although Rad51 and 53BP1 were recruited to these sites. H2AX was dramatically stabilized in response to DSBs directly caused by γ-rays, enabling γH2AX foci formation and DSB repair, whereas H2AX was barely stabilized in response to secondary DSBs, in which γH2AX foci were small and DSBs were not efficiently repaired. Our results show a pathway that leads to the persistent DSB formation after γ-irradiation.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Replication/genetics , Histones/genetics , Rad51 Recombinase/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , 3T3 Cells , Animals , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA Replication/radiation effects , Fibroblasts , Gamma Rays , Gene Expression Regulation/radiation effects , Histones/biosynthesis , Humans , Mice , Rad51 Recombinase/biosynthesis , Tumor Suppressor p53-Binding Protein 1/biosynthesis
14.
Biochem Pharmacol ; 105: 91-100, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26921637

ABSTRACT

Astaxanthin has been demonstrated to exhibit a wide range of beneficial effects, including anti-inflammatory and anti-cancer properties. However, the molecular mechanism of astaxanthin-induced cytotoxicity in non-small cell lung cancer (NSCLC) cells has not been identified. Rad51 plays a central role in homologous recombination, and studies show that chemo-resistant carcinomas exhibit high levels of Rad51 expression. In this study, astaxanthin treatment inhibited cell viability and proliferation of two NSCLC cells, A549 and H1703. Astaxanthin treatment (2.5-20 µM) decreased Rad51 expression and phospho-AKT(Ser473) protein level in a time and dose-dependent manner. Furthermore, expression of constitutively active AKT (AKT-CA) vector rescued the decreased Rad51 mRNA and protein levels in astaxanthin-treated NSCLC cells. Combined treatment with phosphatidylinositol 3-kinase (PI3K) inhibitors (LY294002 or wortmannin) further decreased the Rad51 expression in astaxanthin-exposed A549 and H1703 cells. Knockdown of Rad51 expression by transfection with si-Rad51 RNA or cotreatment with LY294002 further enhanced the cytotoxicity and cell growth inhibition of astaxanthin. Additionally, mitomycin C (MMC) as an anti-tumor antibiotic is widely used in clinical NSCLC chemotherapy. Combination of MMC and astaxanthin synergistically resulted in cytotoxicity and cell growth inhibition in NSCLC cells, accompanied with reduced phospho-AKT(Ser473) level and Rad51 expression. Overexpression of AKT-CA or Flag-tagged Rad51 reversed the astaxanthin and MMC-induced synergistic cytotoxicity. In contrast, pretreatment with LY294002 further decreased the cell viability in astaxanthin and MMC co-treated cells. In conclusion, astaxanthin enhances MMC-induced cytotoxicity by decreasing Rad51 expression and AKT activation. These findings may provide rationale to combine astaxanthin with MMC for the treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Mitomycin/toxicity , Oncogene Protein v-akt/metabolism , Rad51 Recombinase/biosynthesis , Antibiotics, Antineoplastic/toxicity , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Humans , Lung Neoplasms/drug therapy , Mitomycin/therapeutic use , Oncogene Protein v-akt/antagonists & inhibitors , Rad51 Recombinase/antagonists & inhibitors , Xanthophylls/toxicity
15.
Carcinogenesis ; 37(1): 63-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586793

ABSTRACT

RECQL5 is a member of the RecQ family of DNA helicases and has key roles in homologous recombination, base excision repair, replication and transcription. The clinicopathological significance of RECQL5 expression in breast cancer is unknown. In this study, we have evaluated RECQL5 mRNA expression in 1977 breast cancers, and RECQL5 protein level in 1902 breast cancers [Nottingham Tenovus series (n = 1650) and ER- cohort (n = 252)]. Expression levels were correlated to aggressive phenotypes and survival outcomes. High RECQL5 mRNA expression was significantly associated with high histological grade (P = 0.007), HER2 overexpression (P = 0.032), ER+/HER2-/high proliferation genefu subtype (P < 0.0001), integrative molecular clusters (intClust 1and 9) (P < 0.0001) and poor survival (P < 0.0001). In subgroup analysis, high RECQL5 mRNA level remains significantly associated with poor BCSS in ER+ cohort (P < 0.0001) but not in ER- cohort (P = 0.116). At the protein level, in tumours with low RAD51, high RECQL5 level was significantly associated with high histological grade (P < 0.0001), higher mitotic index (P = 0.008), dedifferentiation (P = 0.025), pleomorphism (P = 0.027) and poor survival (P = 0.003). In subgroup analysis, high RECQL5/low RAD51 remains significantly associated with poor BCSS in ER+ cohort (P = 0.010), but not in ER- cohort (P = 0.628). In multivariate analysis, high RECQL5 mRNA and high RECQL5/low RAD51 nuclear protein coexpression independently influenced survival (P = 0.022) in whole cohort and in the ER+ subgroup. Preclinically, we show that exogenous expression of RECQL5 in MCF10A cells can drive proliferation supporting an oncogenic function for RECQL5 in breast cancer. We conclude that RECQL5 is a promising biomarker in breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , RecQ Helicases/biosynthesis , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , DNA Topoisomerases, Type II/biosynthesis , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , MCF-7 Cells , Neoplasm Invasiveness , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , RecQ Helicases/genetics , Tissue Array Analysis
16.
Oncol Rep ; 35(3): 1349-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26676960

ABSTRACT

DNA intrastrand cross-linking agents such as oxaliplatin induce DNA double-strand breaks (DSBs) during DNA repair and replication. In the present study, we hypothesized that DNA intrastrand cross-linking agents may significantly benefit colorectal cancer patients with deficiencies in DSB repair. Seventy-eight patients with metastatic or recurrent colorectal cancer who had measurable target lesions and who underwent resection for primary colorectal cancer in our institution between April 2007 and March 2013 were included in the present study. The median age was 64.5 years, and the cohort consisted of 49 males and 29 females. The median progression-free survival (PFS) was 10.9 months. The expression of DSB repair proteins such as RAD51 and MRE11 was investigated by immunohistochemistry, and associations between RAD51 and MRE11 expression and clinicopathological factors or chemotherapeutic effect were assessed. MRE11-negative cases and RAD51-negative cases achieved significantly better tumor reduction compared with cases with positive expression. Cases with negative expression of both proteins or negative expression of either protein had significantly longer PFS than cases with positive expression for both proteins. In conclusion, DSB repair protein expression-negative colorectal cancer cases may be more highly sensitive to chemotherapy, and thus DSB repair protein expression may be a useful prognostic indicator for colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/drug therapy , DNA Breaks, Double-Stranded/drug effects , DNA-Binding Proteins/biosynthesis , Neoplasm Recurrence, Local/drug therapy , Rad51 Recombinase/biosynthesis , Adult , Aged , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MRE11 Homologue Protein , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prognosis , Rad51 Recombinase/genetics
17.
Tsitologiia ; 58(12): 908-15, 2016.
Article in English, Russian | MEDLINE | ID: mdl-30188106

ABSTRACT

Mouse embryonal fibroblasts with knockout of CDKN1A gene encoding p21/Waf1 protein transformed by oncogenes E1A and cHa-ras (mEras-Waf1­/­ cell line) have been used to assess the level of DNA repair genes expression ­ Rad51 and XRCC5 after treatment with HDAC inhibitor sodium butyrate as compared with their control counterparts (mEras-Waf1+/+ cells). mEras-Waf1­/­ cells are characterized by the elevated amount of single-stranded DNA breaks and g-H2A.X histone foci associated with these breaks. According to immunofluorescence and immunobloting data, Rad51 and Ku80 proteins are highly expressed in the nuclei of both studied cell lines. The level of Ku80 is higher in cells with CDKN1A gene knockout. When cells were treated with DNA-damaging agent adriamycin, there was an additional accumulation of Rad51 foci in the nuclei. However, sodium butyrate reduced considerably the content of Rad51 and Ku80 proteins both in mEras-Waf1+/+ and mEras-Waf1­/­ cells as well as in the cells treated by adriamycin. RT-PCR and immunobloting data show that inhibitory effect of sodium butyrate takes place at the level of Rad51 and XRCC5 gene transcription and the content of Rad51 and Ku80 proteins. The observed suppressive effect of HDACI on DNA repair components explains in part the mechanisms of antiproliferative function of HDAC inhibitors. Surprisingly, sodium butyrate was shown to activate the pluripotent genes transcription in mEras-Waf1+/+ and mEras-Waf1­/­ cells, as exemplified by upregulation of Oct-4, Sox-2, Klf4, implying that these pluripotent genes are under negative control at the level of chromatin structure.


Subject(s)
Butyric Acid/pharmacology , DNA Repair/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Ku Autoantigen/biosynthesis , Rad51 Recombinase/biosynthesis , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , DNA Repair/genetics , Kruppel-Like Factor 4 , Ku Autoantigen/genetics , Mice , Rad51 Recombinase/genetics , Transcription, Genetic/drug effects
18.
Oncotarget ; 6(27): 24474-87, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26090616

ABSTRACT

DNA polymerase beta (Pol ß) is a key enzyme for the protection against oxidative DNA lesions via its role in base excision repair (BER). Approximately 1/3 of tumors studied to date express Pol ß variant proteins, and several tumors overexpress Pol ß. Pol ß possesses DNA polymerase and dRP lyase activities, both of which are known to be important for efficient BER. The dRP lyase activity resides within the 8kDa amino terminal domain of Pol ß, is responsible for removal of the 5' phosphate group (5'-dRP). The DNA polymerase subsequently fills the gaps. Previously, we demonstrated that the human gastric cancer-associated variant of Pol ß (Leu22Pro (L22P)) lacks dRP lyase function in vitro. Here, we report that L22P-expressing cells harbor significantly increased replication associated DNA double strand breaks (DSBs) and defective maintenance of the nascent DNA strand (NDS) during replication stress. Moreover, L22P-expressing cells are sensitive to PARP1 inhibitors, which suggests trapped PARP1 binds to the 5'-dRP group and blocks replications forks, resulting in fork collapse and DSBs. Our data suggest that the normal function of the dRP lyase is critical to maintain replication fork integrity and prevent replication fork collapse to DSBs and cellular transformation.


Subject(s)
DNA Breaks, Double-Stranded , DNA Polymerase beta/genetics , DNA Repair/genetics , DNA Replication/genetics , Stomach Neoplasms/genetics , Cell Line, Tumor , DNA, Single-Stranded/genetics , Humans , Phosphorus-Oxygen Lyases/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Isoforms/genetics , Rad51 Recombinase/biosynthesis
19.
Cell Res ; 25(3): 351-69, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25698579

ABSTRACT

The inhibitory role of p53 in DNA double-strand break (DSB) repair seems contradictory to its tumor-suppressing property. The p53 isoform Δ113p53/Δ133p53 is a p53 target gene that antagonizes p53 apoptotic activity. However, information on its functions in DNA damage repair is lacking. Here we report that Δ113p53 expression is strongly induced by γ-irradiation, but not by UV-irradiation or heat shock treatment. Strikingly, Δ113p53 promotes DNA DSB repair pathways, including homologous recombination, non-homologous end joining and single-strand annealing. To study the biological significance of Δ113p53 in promoting DNA DSB repair, we generated a zebrafish Δ113p53(M/M) mutant via the transcription activator-like effector nuclease technique and found that the mutant is more sensitive to γ-irradiation. The human ortholog, Δ133p53, is also only induced by γ-irradiation and functions to promote DNA DSB repair. Δ133p53-knockdown cells were arrested at the G2 phase at the later stage in response to γ-irradiation due to a high level of unrepaired DNA DSBs, which finally led to cell senescence. Furthermore, Δ113p53/Δ133p53 promotes DNA DSB repair via upregulating the transcription of repair genes rad51, lig4 and rad52 by binding to a novel type of p53-responsive element in their promoters. Our results demonstrate that Δ113p53/Δ133p53 is an evolutionally conserved pro-survival factor for DNA damage stress by preventing apoptosis and promoting DNA DSB repair to inhibit cell senescence. Our data also suggest that the induction of Δ133p53 expression in normal cells or tissues provides an important tolerance marker for cancer patients to radiotherapy.


Subject(s)
Apoptosis/genetics , Cellular Senescence/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , Tumor Suppressor Protein p53/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Cell Line , DNA/genetics , DNA Ligase ATP , DNA Ligases/biosynthesis , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/radiation effects , Humans , Promoter Regions, Genetic/genetics , Protein Binding , Protein Isoforms/genetics , RNA Interference , RNA, Small Interfering , Rad51 Recombinase/biosynthesis , Zebrafish , Zebrafish Proteins/biosynthesis
20.
J Cell Sci ; 128(2): 317-30, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25395584

ABSTRACT

DNA double-strand breaks (DSBs) are a type of lethal DNA damage. The repair of DSBs requires tight coordination between the factors modulating chromatin structure and the DNA repair machinery. BRG1, the ATPase subunit of the chromatin remodelling complex Switch/Sucrose non-fermentable (SWI/SNF), is often linked to tumorigenesis and genome instability, and its role in DSB repair remains largely unclear. In the present study, we show that BRG1 is recruited to DSB sites and enhances DSB repair. Using DR-GFP and EJ5-GFP reporter systems, we demonstrate that BRG1 facilitates homologous recombination repair rather than nonhomologous end-joining (NHEJ) repair. Moreover, the BRG1-RAD52 complex mediates the replacement of RPA with RAD51 on single-stranded DNA (ssDNA) to initiate DNA strand invasion. Loss of BRG1 results in a failure of RAD51 loading onto ssDNA, abnormal homologous recombination repair and enhanced DSB-induced lethality. Our present study provides a mechanistic insight into how BRG1, which is known to be involved in chromatin remodelling, plays a substantial role in the homologous recombination repair pathway in mammalian cells.


Subject(s)
DNA End-Joining Repair/genetics , DNA Helicases/genetics , Nuclear Proteins/genetics , Rad51 Recombinase/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Replication Protein A/genetics , Transcription Factors/genetics , Cell Line , Chromatin Assembly and Disassembly/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , DNA Damage/genetics , DNA Helicases/biosynthesis , DNA, Single-Stranded/drug effects , DNA, Single-Stranded/genetics , Etoposide/toxicity , Genomic Instability , Homologous Recombination/genetics , Humans , Nuclear Proteins/biosynthesis , Rad51 Recombinase/biosynthesis , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...