Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
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.
J Biol Chem ; 296: 100355, 2021.
Article in English | MEDLINE | ID: mdl-33539926

ABSTRACT

The MCM8/9 complex is implicated in aiding fork progression and facilitating homologous recombination (HR) in response to several DNA damage agents. MCM9 itself is an outlier within the MCM family containing a long C-terminal extension (CTE) comprising 42% of the total length, but with no known functional components and high predicted disorder. In this report, we identify and characterize two unique motifs within the primarily unstructured CTE that are required for localization of MCM8/9 to sites of mitomycin C (MMC)-induced DNA damage. First, an unconventional "bipartite-like" nuclear localization (NLS) motif consisting of two positively charged amino acid stretches separated by a long intervening sequence is required for the nuclear import of both MCM8 and MCM9. Second, a variant of the BRC motif (BRCv) similar to that found in other HR helicases is necessary for localization to sites of MMC damage. The MCM9-BRCv directly interacts with and recruits RAD51 downstream to MMC-induced damage to aid in DNA repair. Patient lymphocytes devoid of functional MCM9 and discrete MCM9 knockout cells have a significantly impaired ability to form RAD51 foci after MMC treatment. Therefore, the disordered CTE in MCM9 is functionally important in promoting MCM8/9 activity and in recruiting downstream interactors; thus, requiring full-length MCM9 for proper DNA repair.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA Damage/drug effects , Minichromosome Maintenance Proteins/metabolism , Mitomycin/pharmacology , Rad51 Recombinase/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Minichromosome Maintenance Proteins/analysis , Rad51 Recombinase/analysis
3.
Nucleic Acids Res ; 49(4): 2005-2026, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33476370

ABSTRACT

Replication Protein A (RPA) is a critical complex that acts in replication and promotes homologous recombination by allowing recombinase recruitment to processed DSB ends. Most organisms possess three RPA subunits (RPA1, RPA2, RPA3) that form a trimeric complex critical for viability. The Caenorhabditis elegans genome encodes RPA-1, RPA-2 and an RPA-2 paralog RPA-4. In our analysis, we determined that RPA-2 is critical for germline replication and normal repair of meiotic DSBs. Interestingly, RPA-1 but not RPA-2 is essential for somatic replication, in contrast to other organisms that require both subunits. Six different hetero- and homodimeric complexes containing permutations of RPA-1, RPA-2 and RPA-4 can be detected in whole animal extracts. Our in vivo studies indicate that RPA-1/4 dimer is less abundant in the nucleus and its formation is inhibited by RPA-2. While RPA-4 does not participate in replication or recombination, we find that RPA-4 inhibits RAD-51 filament formation and promotes apoptosis of a subset of damaged nuclei. Altogether these findings point to sub-functionalization and antagonistic roles of RPA complexes in C. elegans.


Subject(s)
Apoptosis , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , DNA Replication , Meiosis/genetics , Recombination, Genetic , Replication Protein A/physiology , Animals , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/metabolism , DNA Breaks, Double-Stranded , Mitosis/genetics , Rad51 Recombinase/analysis , Replication Protein A/metabolism
4.
J Clin Pathol ; 74(4): 223-227, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32220941

ABSTRACT

AIMS: The aim of this study is to investigate the expression profiles of cell cycle related proteins in nasal extranodal NK/T cell lymphoma, nasal type (ENKTCL). METHODS: The expression profiles of cell cycle related proteins were assessed with a cell cycle antibody array and validated by immunohistochemistry. Correlations between the expression levels of proteins and clinical outcomes of patients with nasal ENKTCL were evaluated. RESULTS: The expression of full length ataxia telangiectasia mutated (ATM) in nasal ENKTCL significantly decreased compared with that in nasal benign lymphoid proliferative disease (NBLPD), but the expression levels of p-ATM, CHK2 and RAD51 significantly increased in nasal ENKTCL compared with that in NBLPD. Kaplan-Meier analysis showed that the expression levels of p-ATM and CHK2 in nasal ENKTCL were inversely related to overall survival (p=0.011 and p=0.025, respectively). CONCLUSION: Abnormalities in the ATM pathway may play a crucial role in the oncogenesis and chemoradiotherapy resistance of nasal ENKTCL.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/analysis , Biomarkers, Tumor/analysis , Checkpoint Kinase 2/analysis , Lymphoma, Extranodal NK-T-Cell/enzymology , Cell Proliferation , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Lymphoma, Extranodal NK-T-Cell/mortality , Lymphoma, Extranodal NK-T-Cell/pathology , Lymphoma, Extranodal NK-T-Cell/therapy , Neoplasm Grading , Phosphorylation , Rad51 Recombinase/analysis , Radiation Tolerance , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
5.
Int J Radiat Oncol Biol Phys ; 108(3): 758-769, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32407930

ABSTRACT

PURPOSE: Tumor resistance to radiation therapy is a therapeutic challenge in the treatment of patients with non-small cell lung cancer. Cyclin-dependent kinase 5 (CDK5) has been proposed to participate in cell proliferation, migration and invasion, drug resistance, and immune evasion. However, the functions and regulatory mechanisms of CDK5 in lung cancer radioresistance have not been investigated. METHODS AND MATERIALS: DNA damage response and repair were measured by neutral comet assay and γ-H2AX and Rad51 foci staining. The biological functions of CDK5 in lung cancer radioresistance were investigated with clonogenic survival assays and xenograft tumor models. Small interfering RNAs and short hairpin RNAs were used to knock down CDK5 in A549 and H1299 cells. The effects of CDK5 depletion on the tumorigenic behaviors of lung cancer cells were evaluated in vitro and in vivo. Gene expression was examined by RNA-seq and quantitative real-time polymerase chain reaction. RESULTS: We report that CDK5 depletion impairs lung cancer progression and radioresistance in vitro and in vivo. Mechanistically, we identify TAZ, a component of the Hippo pathway, as a critical downstream effector of CDK5. Loss of CDK5 downregulates TAZ expression and attenuates Hippo signaling activation. Importantly, we provide evidence that TAZ is the major effector mediating the biological functions of CDK5 in lung cancer. CONCLUSIONS: These results illustrate that CDK5 activates Hippo signaling via TAZ to participate in tumorigenesis and radioresistance, suggesting that CDK5 may be a promising radiosensitization target for the treatment of lung cancer.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Lung Neoplasms/radiotherapy , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/physiology , Transcription Factors/metabolism , A549 Cells , Acyltransferases , Animals , Cell Line, Tumor , Comet Assay , Cyclin-Dependent Kinase 5/deficiency , Cyclin-Dependent Kinase 5/genetics , DNA Damage , DNA Repair , Disease Progression , Down-Regulation , Fluorescent Antibody Technique , Gene Knockdown Techniques , Gene Silencing , Heterografts , Hippo Signaling Pathway , Histones/analysis , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering , Rad51 Recombinase/analysis , Up-Regulation
6.
Anal Cell Pathol (Amst) ; 2020: 1827676, 2020.
Article in English | MEDLINE | ID: mdl-32190537

ABSTRACT

PURPOSE: To examine the expression of RAD51 in oral squamous cell carcinoma (OSCC) and analyze its connection with pathological grade, clinical stage, and lymphatic metastasis potential. METHODS: For this study, 74 OSCC samples, 15 normal mucosa tissues, and 11 normal skin tissue samples were collected. RAD51 expression was investigated using immunohistochemistry. A follow-up visit was used to assess the prognosis of each patient. We compared RAD51 expression in oral mucosa epithelial cells (OMECs), keratinocytes, and tongue squamous cell carcinoma cells (TSCCs) by Western blot analysis. RESULTS: RAD51 expression was higher in tumor cells than in normal mucosal tissues. In addition, RAD51 expression was associated with higher tumor differentiation (P < 0.05). Also, RAD51 expression was higher (P < 0.05). Also, RAD51 expression was higher (P < 0.05). Also, RAD51 expression was higher (. CONCLUSION: A strong positive correlation was found between RAD51 expression and the degree of malignancy in OSCC patients, suggesting that RAD51 could be an excellent prognostic indicator for OSCC patients.


Subject(s)
Biomarkers, Tumor/analysis , Mouth Neoplasms/pathology , Rad51 Recombinase/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Rad51 Recombinase/analysis
7.
Medicina (Kaunas) ; 56(2)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973027

ABSTRACT

Background and Objectives: RAD51 plays an essential role in DNA repair via homologous recombination. RAD51 facilitates strand transfer between interrupted sequences and their undamaged homologies. Therefore, we studied the RAD51 mRNA expression levels in colorectal cancer (CRC), and evaluated the clinicopathological and prognostic significance of RAD51. Materials and Methods: The RAD51 expression was examined in 48 CRCs and paired adjacent non-tumor tissues. We further evaluated the survival to determine the prognostic value of RAD51 in our CRC and The Cancer Genome Atlas (TCGA) data. Results: We confirmed that the RAD51 expression in tumor tissues, compared with that of paired non-tumor tissues, was upregulated 2.5-fold. Additionally, the RAD51 expression was significantly associated with the T stage (p = 0.027). According to a higher T stage, the RAD51 expression showed an increasing trend. However, the RAD51 expression did not show a prognostic value statistically. Conclusions: We confirmed that RAD51 was upregulated in tumors and was significantly associated with the T stage. Although there was no statistically significant prognostic value found in our samples and TGCA data, our study will provide new insight for RAD51 in CRC.


Subject(s)
Colorectal Neoplasms/blood , Predictive Value of Tests , Rad51 Recombinase/analysis , Aged , Chi-Square Distribution , Colorectal Neoplasms/genetics , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Rad51 Recombinase/blood , Rad51 Recombinase/genetics , Survival Analysis
8.
Gynecol Oncol ; 152(1): 157-165, 2019 01.
Article in English | MEDLINE | ID: mdl-30414739

ABSTRACT

OBJECTIVE: Poly(ADP-ribose) polymerase inhibitors (PARPi) are active in cancer cells that have impaired repair of DNA by the homologous recombination (HR) pathway. Strategies that disrupt HR may sensitize HR-proficient tumors to PARP inhibition. As a component of the core cell cycle machinery, cyclin D1 has unexpected function in DNA repair, suggesting that targeting cyclin D1 may represent a plausible strategy for expanding the utility of PARPi in ovarian cancer. METHODS: BRCA1 wildtype ovarian cancer cells (A2780 and SKOV3) were treated with a combination of CCND1 siRNA and olaparib in vitro. Cell viability was assessed by MTT. The effects of the combined treatment on DNA damage repair and cell cycle progression were examined to dissect molecular mechanisms. In vivo studies were performed in an orthotopic ovarian cancer mouse model. Animals were treated with a combination of lentivirus-mediated CCND1 shRNA and olaparib or olaparib plus scrambled shRNA. Molecular downstream effects were examined by immunohistochemistry. RESULTS: Silencing of cyclin D1 sensitized ovarian cancer cells to olaparib through interfering with RAD51 accumulation and inducing cell cycle G0/G1 arrest. Treatment of lentivirus-mediated CCND1-shRNA in nude mice statistically significantly augmented the olaparib response (mean tumor weight ±â€¯SD, CCND1-shRNA plus olaparib vs scrambled shRNA plus olaparib: 0.172 ±â€¯0.070 g vs 0.324 ±â€¯0.044 g, P< 0.05). CONCLUSIONS: Silencing of cyclin D1 combined with olaparib may lead to substantial benefit for ovarian cancer management by mimicking a BRCAness phenotype, and induction of G0/G1 cell cycle arrest.


Subject(s)
Cyclin D1/physiology , DNA Breaks, Double-Stranded , DNA Repair , Genes, BRCA1 , Ovarian Neoplasms/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Animals , Cell Cycle , Cell Line, Tumor , Cyclin D1/genetics , Female , Humans , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Rad51 Recombinase/analysis
9.
EMBO Mol Med ; 10(12)2018 12.
Article in English | MEDLINE | ID: mdl-30377213

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are effective in cancers with defective homologous recombination DNA repair (HRR), including BRCA1/2-related cancers. A test to identify additional HRR-deficient tumors will help to extend their use in new indications. We evaluated the activity of the PARPi olaparib in patient-derived tumor xenografts (PDXs) from breast cancer (BC) patients and investigated mechanisms of sensitivity through exome sequencing, BRCA1 promoter methylation analysis, and immunostaining of HRR proteins, including RAD51 nuclear foci. In an independent BC PDX panel, the predictive capacity of the RAD51 score and the homologous recombination deficiency (HRD) score were compared. To examine the clinical feasibility of the RAD51 assay, we scored archival breast tumor samples, including PALB2-related hereditary cancers. The RAD51 score was highly discriminative of PARPi sensitivity versus PARPi resistance in BC PDXs and outperformed the genomic test. In clinical samples, all PALB2-related tumors were classified as HRR-deficient by the RAD51 score. The functional biomarker RAD51 enables the identification of PARPi-sensitive BC and broadens the population who may benefit from this therapy beyond BRCA1/2-related cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/diagnosis , Drug Resistance, Neoplasm , Heterografts/pathology , Phthalazines/administration & dosage , Piperazines/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Rad51 Recombinase/analysis , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Homologous Recombination , Humans , Mice
10.
Nucleic Acids Res ; 46(12): 6238-6256, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29788478

ABSTRACT

Fanconi Anemia (FA) is characterized by bone marrow failure, congenital abnormalities, and cancer. Of over 20 FA-linked genes, FANCJ uniquely encodes a DNA helicase and mutations are also associated with breast and ovarian cancer. fancj-/- cells are sensitive to DNA interstrand cross-linking (ICL) and replication fork stalling drugs. We delineated the molecular defects of two FA patient-derived FANCJ helicase domain mutations. FANCJ-R707C was compromised in dimerization and helicase processivity, whereas DNA unwinding by FANCJ-H396D was barely detectable. DNA binding and ATP hydrolysis was defective for both FANCJ-R707C and FANCJ-H396D, the latter showing greater reduction. Expression of FANCJ-R707C or FANCJ-H396D in fancj-/- cells failed to rescue cisplatin or mitomycin sensitivity. Live-cell imaging demonstrated a significantly compromised recruitment of FANCJ-R707C to laser-induced DNA damage. However, FANCJ-R707C expressed in fancj-/- cells conferred resistance to the DNA polymerase inhibitor aphidicolin, G-quadruplex ligand telomestatin, or DNA strand-breaker bleomycin, whereas FANCJ-H396D failed. Thus, a minimal threshold of FANCJ catalytic activity is required to overcome replication stress induced by aphidicolin or telomestatin, or to repair bleomycin-induced DNA breakage. These findings have implications for therapeutic strategies relying on DNA cross-link sensitivity or heightened replication stress characteristic of cancer cells.


Subject(s)
DNA Breaks, Double-Stranded , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , DNA Replication , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Aphidicolin/toxicity , Cell Line , Checkpoint Kinase 1/metabolism , Chickens , Cisplatin/toxicity , DNA, Single-Stranded , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins/chemistry , G-Quadruplexes , Mutation, Missense , Oxazoles/toxicity , RNA Helicases/chemistry , Rad51 Recombinase/analysis , Recombinases/genetics , Recombinases/metabolism , Replication Protein A/metabolism , Stress, Physiological
11.
Biochemistry (Mosc) ; 83(1): 69-75, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29534671

ABSTRACT

Colon cancer remains one of the most common digestive system malignancies in the World. This study investigated the possible interaction between RAD51 and minichromosome maintenance proteins (MCMs) in HCT116 cells, which can serve as a model system for forming colon cancer foci. The interaction between RAD51 and MCMs was detected by mass spectrometry. Silenced MCM vectors were transfected into HTC116 cells. The expressions of RAD51 and MCMs were detected using Western blotting. Foci forming and chromatin fraction of RAD51 in HCT116 cells were also analyzed. The results showed that RAD51 directly interacted with MCM2, MCM3, MCM5, and MCM6 in colon cancer HTC116 cells. Suppression of MCM2 or MCM6 by shRNA decreased the chromatin localization of RAD51 in HTC116 cells. Moreover, silenced MCM2 or MCM6 decreased the foci forming of RAD51 in HTC116 cells. Our study suggests that the interaction between MCMs and RAD51 is essential for the chromatin localization and foci forming of RAD51 in HCT116 cell DNA damage recovery, and it may be a theoretical basis for analysis of RAD51 in tumor samples of colon cancer patients.


Subject(s)
Colonic Neoplasms/metabolism , Minichromosome Maintenance Proteins/metabolism , Rad51 Recombinase/metabolism , Cells, Cultured , Colonic Neoplasms/chemistry , HCT116 Cells , HEK293 Cells , Humans , Mass Spectrometry , Minichromosome Maintenance Proteins/analysis , Minichromosome Maintenance Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Rad51 Recombinase/analysis , Rad51 Recombinase/antagonists & inhibitors
12.
Methods Enzymol ; 600: 201-232, 2018.
Article in English | MEDLINE | ID: mdl-29458759

ABSTRACT

Human RAD51 promotes accurate DNA repair by homologous recombination and is involved in protection and repair of damaged DNA replication forks. The active species of RAD51 and related recombinases in all organisms is a nucleoprotein filament assembled on single-stranded DNA (ssDNA). The formation of a nucleoprotein filament competent for the recombination reaction, or for DNA replication support, is a delicate and strictly regulated process, which occurs through filament nucleation followed by filament extension. The rates of these two phases of filament formation define the capacity of RAD51 to compete with the ssDNA-binding protein RPA, as well as the lengths of the resulting filament segments. Single-molecule approaches can provide a wealth of quantitative information on the kinetics of RAD51 nucleoprotein filament assembly, internal dynamics, and disassembly. In this chapter, we describe how to set up a single-molecule total internal reflection fluorescence microscopy experiment to monitor the initial steps of RAD51 nucleoprotein filament formation in real-time and at single-monomer resolution. This approach is based on the unique, stretched-ssDNA conformation within the recombinase nucleoprotein filament and follows the efficiency of Förster resonance energy transfer (EFRET) between two DNA-conjugated fluorophores. We will discuss the practical aspects of the experimental setup, extraction of the FRET trajectories, and how to analyze and interpret the data to obtain information on RAD51 nucleation kinetics, the mechanism of nucleation, and the oligomeric species involved in filament formation.


Subject(s)
DNA, Single-Stranded/metabolism , Nucleoproteins/analysis , Rad51 Recombinase/analysis , Recombinational DNA Repair , Single Molecule Imaging/methods , DNA, Single-Stranded/chemistry , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Humans , Kinetics , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Molecular Conformation , Nucleoproteins/chemistry , Nucleoproteins/metabolism , Protein Binding , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Single Molecule Imaging/instrumentation
13.
Methods Enzymol ; 600: 375-406, 2018.
Article in English | MEDLINE | ID: mdl-29458767

ABSTRACT

Direct observation of individual protein molecules in their native environment, at nanometer resolution, in a living cell, in motion is not only fascinating but also uniquely informative. Several recent major technological advances in genomic engineering, protein and synthetic fluorophore development, and light microscopy have dramatically increased the accessibility of this approach. This chapter describes the procedures for modifying endogenous genomic loci to producing fluorescently tagged proteins, their high-resolution visualization, and analysis of their dynamics in mammalian cells, using DNA repair proteins BRCA2 and RAD51 as an example.


Subject(s)
BRCA2 Protein/analysis , Cell Culture Techniques/methods , Intravital Microscopy/methods , Rad51 Recombinase/analysis , Recombinational DNA Repair , Single Molecule Imaging/methods , Animals , BRCA2 Protein/chemistry , BRCA2 Protein/metabolism , CRISPR-Cas Systems/genetics , Cell Culture Techniques/instrumentation , Cells, Cultured , DNA Breaks, Double-Stranded , Fluorescence Recovery After Photobleaching/instrumentation , Fluorescence Recovery After Photobleaching/methods , Gene Editing/methods , Green Fluorescent Proteins/chemistry , Intravital Microscopy/instrumentation , Luminescent Agents/chemistry , Mice , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Mouse Embryonic Stem Cells , Protein Binding , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Single Molecule Imaging/instrumentation
14.
Nucleic Acids Res ; 46(5): 2432-2445, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29346668

ABSTRACT

Replicative erosion of telomeres is naturally compensated by telomerase and studies in yeast and vertebrates show that homologous recombination can compensate for the absence of telomerase. We show that RAD51 protein, which catalyzes the key strand-invasion step of homologous recombination, is localized at Arabidopsis telomeres in absence of telomerase. Blocking the strand-transfer activity of the RAD51 in telomerase mutant plants results in a strikingly earlier onset of developmental defects, accompanied by increased numbers of end-to-end chromosome fusions. Imposing replication stress through knockout of RNaseH2 increases numbers of chromosome fusions and reduces the survival of these plants deficient for telomerase and homologous recombination. This finding suggests that RAD51-dependent homologous recombination acts as an essential backup to the telomerase for compensation of replicative telomere loss to ensure genome stability. Furthermore, we show that this positive role of RAD51 in telomere stability is dependent on the RTEL1 helicase. We propose that a RAD51 dependent break-induced replication process is activated in cells lacking telomerase activity, with RTEL1 responsible for D-loop dissolution after telomere replication.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/genetics , DNA Helicases/physiology , Rad51 Recombinase/physiology , Telomere Shortening , Arabidopsis/enzymology , Arabidopsis Proteins/analysis , DNA Replication , Genomic Instability , Homologous Recombination , Mutation , Rad51 Recombinase/analysis , Repetitive Sequences, Nucleic Acid , Ribonucleases/genetics , Stochastic Processes , Telomerase/genetics , Telomere/chemistry
15.
Med Microbiol Immunol ; 206(4): 327-335, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28466382

ABSTRACT

Adult T cell leukemia/lymphoma (ATLL) is a life-threatening malignancy of HTLV-1 infected Th lymphocytes. In the present study host-virus interactions were investigated by assessment of HTLV-1 proviral load (PVL) and host gene expression. A cross-sectional study was carried out on 18 ATLL, 10 HAM/TSP patients and 18 HTLV-1 asymptomatic carriers (ACs). DNA and mRNA of the peripheral blood mononuclear cells were extracted for PVL and LAT, BIM, c-FOS and RAD51 gene expression measurement using qRT-PCR. The mean PVL in ATLL patients was 11,430 ± 3770 copies/104 which was statistically higher than ACs, 530 ± 119 copies/104, (p < 0.001). The expression of BIM, and c-FOS in ATLL patients were higher than HTLV-1 ACs; however, there were no statistically significant differences. The expression of RAD51 as an essential player on DNA repair showed around 160 times increase in ATLL group (166 ± 95) compared to ACs (1.04 ± 0.34) which is statistically significant (p < 0.001). Interestingly, there was a positive correlation between RAD51 expression and HTLV-PVL. The expression of LAT as a central adaptor in TCR signaling interestingly was around 36 times higher in ATLL group than ACs (ATLL; 41.33 ± 19.91 vs. ACs; 1.15 ± 0.22, p < 0.001). This finding showed that TCR signaling pathway mainly provides the growth factors for transformed cells. Furthermore, the overexpression of RAD51 which has been induced in HTLV-1 infected cells as a consequence of virus replication is not able to overcome the DNA damage toward cell transformation.


Subject(s)
Adaptor Proteins, Signal Transducing/analysis , Bcl-2-Like Protein 11/analysis , Human T-lymphotropic virus 1/isolation & purification , Leukemia-Lymphoma, Adult T-Cell/pathology , Membrane Proteins/analysis , Proto-Oncogene Proteins c-fos/analysis , Rad51 Recombinase/analysis , Viral Load , Adult , Cross-Sectional Studies , Female , Gene Expression Profiling , Humans , Leukemia-Lymphoma, Adult T-Cell/virology , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/virology , Male , Middle Aged , Proviruses/isolation & purification
16.
Int J Radiat Oncol Biol Phys ; 95(2): 772-81, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27020103

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase (PARP) inhibitors potentiate radiation therapy in preclinical models of human non-small cell lung cancer (NSCLC) and other types of cancer. However, the mechanisms underlying radiosensitization in vivo are incompletely understood. Herein, we investigated the impact of hypoxia on radiosensitization by the PARP inhibitor olaparib in human NSCLC xenograft models. METHODS AND MATERIALS: NSCLC Calu-6 and Calu-3 cells were irradiated in the presence of olaparib or vehicle under normoxic (21% O2) or hypoxic (1% O2) conditions. In vitro radiosensitivity was assessed by clonogenic survival assay and γH2AX foci assay. Established Calu-6 and Calu-3 subcutaneous xenografts were treated with olaparib (50 mg/kg, daily for 3 days), radiation (10 Gy), or both. Tumors (n=3/group) were collected 24 or 72 hours after the first treatment. Immunohistochemistry was performed to assess hypoxia (carbonic anhydrase IX [CA9]), vessels (CD31), DNA double strand breaks (DSB) (γH2AX), and apoptosis (cleaved caspase 3 [CC3]). The remaining xenografts (n=6/group) were monitored for tumor growth. RESULTS: In vitro, olaparib showed a greater radiation-sensitizing effect in Calu-3 and Calu-6 cells in hypoxic conditions (1% O2). In vivo, Calu-3 tumors were well-oxygenated, whereas Calu-6 tumors had extensive regions of hypoxia associated with down-regulation of the homologous recombination protein RAD51. Olaparib treatment increased unrepaired DNA DSB (P<.001) and apoptosis (P<.001) in hypoxic cells of Calu-6 tumors following radiation, whereas it had no significant effect on radiation-induced DNA damage response in nonhypoxic cells of Calu-6 tumors or in the tumor cells of well-oxygenated Calu-3 tumors. Consequently, olaparib significantly increased radiation-induced growth inhibition in Calu-6 tumors (P<.001) but not in Calu-3 tumors. CONCLUSIONS: Our data suggest that hypoxia potentiates the radiation-sensitizing effects of olaparib by contextual synthetic killing, and that tumor hypoxia may be a potential biomarker for selecting patients who may get the greatest benefit from the addition of olaparib to radiation therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Phthalazines/pharmacology , Piperazines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia , Cell Line, Tumor , DNA Damage , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Rad51 Recombinase/analysis , Xenograft Model Antitumor Assays
17.
Radiat Res ; 182(5): 475-88, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25361396

ABSTRACT

Hypoxia exists in all solid tumors and leads to clinical radioresistance and adverse prognosis. We hypothesized that hypoxia and cellular localization of gold nanoparticles (AuNPs) could be modifiers of AuNP-mediated radiosensitization. The possible mechanistic effect of AuNPs on cell cycle distribution and DNA double-strand break (DSB) repair postirradiation were also studied. Clonogenic survival data revealed that internalized and extracellular AuNPs at 0.5 mg/mL resulted in dose enhancement factors of 1.39 ± 0.07 and 1.09 ± 0.01, respectively. Radiosensitization by AuNPs was greatest in cells under oxia, followed by chronic and then acute hypoxia. The presence of AuNPs inhibited postirradiation DNA DSB repair, but did not lead to cell cycle synchronization. The relative radiosensitivity of chronic hypoxic cells is attributed to defective DSB repair (homologous recombination) due to decreased (RAD51)-associated protein expression. Our results support the need for further study of AuNPs for clinical development in cancer therapy since their efficacy is not limited in chronic hypoxic cells.


Subject(s)
Breast Neoplasms/radiotherapy , Gold/pharmacology , Metal Nanoparticles/administration & dosage , Radiation-Sensitizing Agents/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Hypoxia , Cell Line, Tumor , DNA Breaks, Double-Stranded , Female , Humans , Rad51 Recombinase/analysis , Reactive Oxygen Species/metabolism
18.
Int J Radiat Oncol Biol Phys ; 89(5): 1106-1114, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25035215

ABSTRACT

PURPOSE: To test the hypothesis that small molecule targeting of nucleophosmin 1 (NPM1) represents a rational approach for radiosensitization. METHODS AND MATERIALS: Wilde-type and NPM1-deficient mouse embryo fibroblasts (MEFs) were used to determine whether radiosensitization produced by the small molecule YTR107 was NPM1 dependent. The stress response to ionizing radiation was assessed by quantifying pNPM1, γH2AX, and Rad51 foci, neutral comet tail moment, and colony formation. NPM1 levels in a human-derived non-small-cell lung cancer (NSCLC) tissue microarray (TMA) were determined by immunohistochemistry. YTR107-mediated radiosensitization was assessed in NSCLC cell lines and xenografts. RESULTS: Use of NPM1-null MEFs demonstrated that NPM1 is critical for DNA double- strand break (DSB) repair, that loss of NPM1 increases radiation sensitivity, and that YTR107-mediated radiosensitization is NPM1 dependent. YTR107 was shown to inhibit NPM1 oligomerization and impair formation of pNPM1 irradiation-induced foci that colocalized with γH2AX foci. Analysis of the TMA demonstrated that NPM1 is overexpressed in subsets of NSCLC. YTR107 inhibited DNA DSB repair and radiosensitized NSCLC lines and xenografts. CONCLUSIONS: These data demonstrate that YTR107-mediated targeting of NPM1 impairs DNA DSB repair, an event that increases radiation sensitivity.


Subject(s)
Barbiturates/pharmacology , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Indoles/pharmacology , Molecular Targeted Therapy/methods , Neoplasm Proteins/drug effects , Nuclear Proteins/drug effects , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/radiotherapy , Fibroblasts/chemistry , Fibroblasts/radiation effects , Histones/analysis , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/analysis , Neoplasm Proteins/deficiency , Neoplasm Proteins/physiology , Nuclear Proteins/analysis , Nuclear Proteins/deficiency , Nuclear Proteins/physiology , Nucleophosmin , Rad51 Recombinase/analysis , Tissue Array Analysis , Tumor Cells, Cultured
19.
Breast ; 22(6): 1178-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24060578

ABSTRACT

BACKGROUND: There are no clinically useful biomarkers predictive of brain metastases (BM) in breast cancer. In this study, we investigated the correlation between expression of selected proteins in the primary tumor and the risk of BM in patients with metastatic breast cancer (MBC). METHODS: The study included 198 MBC patients (96 with and 102 without BM). Using tissue microarrays derived from the primary tumor, we assessed by immunohistochemical expression of ER, PR, HER2, Ki-67, CK5/6, EGFR, HER3, CXCR4, Rad51, E-cadherin, and claudin 3 and 4. RESULTS: Ki-67 ≥14% (hazard ratio [HR] 2.76; P < 0.001), cytoplasmic expression of Rad51 (HR 1.87; P = 0.014) and ER-negativity (HR 1.72; P = 0.029) were associated with increased risk of BM in the multivariate analysis. A three-biomarker profile including ER, Ki-67 and Rad51 vs. other subtypes combined yielded an HR of 4.43 (P < 0.001). CONCLUSIONS: ER-negativity, cytoplasmic expression of Rad51 and high Ki-67 are associated with increased risk of BM.


Subject(s)
Biomarkers, Tumor/analysis , Brain Neoplasms/secondary , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Lobular/chemistry , Rad51 Recombinase/analysis , Adult , Aged , Aged, 80 and over , Cadherins/analysis , Carcinoma, Ductal, Breast/secondary , Carcinoma, Lobular/secondary , Claudin-3/analysis , Claudin-4/analysis , Female , Humans , Immunohistochemistry , Keratin-5/analysis , Keratin-6/analysis , Ki-67 Antigen/analysis , Middle Aged , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis , Receptors, CXCR4/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Risk Factors , Tissue Array Analysis , Young Adult
20.
Radiother Oncol ; 108(3): 429-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23932191

ABSTRACT

BACKGROUND AND PURPOSE: In muscle-invasive bladder cancer there is an urgent need to identify relatively non-toxic radiosensitising agents for use in elderly patients. Histone deacetylase inhibitors radiosensitise tumour cells but not normal cells in vitro and variously downregulate DNA damage signalling, homologous recombination (HR) and non-homologous end-joining (NHEJ) repair proteins. We investigated panobinostat (PAN) as a potential radiosensitiser in bladder cancer cells. MATERIALS AND METHODS: Clonogenic assays were performed in RT112 bladder cancer cells, and RT112 cells stably knocked down for RAD51 or Ku80 by shRNAi. Resolution of γH2AX foci was determined by immunofluorescence confocal microscopy, cell cycle progression by FACS analysis and protein expression by western blotting. RESULTS: PAN had a greater radiosensitising effect in Ku80KD than RT112 or RAD51KD cells; enhancement ratios 1.35 for Ku80KD at 10nM (IC(20) for Ku80KD) and 1.31 for RT112 and RAD51KD at 25 nM (IC(40) for both). PAN downregulated MRE11, NBS1 and RAD51, but not Ku70 and Ku80, increased γH2AX foci formation in a dose-dependent manner and delayed γH2AX foci repair after ionising radiation. CONCLUSIONS: PAN acts as a radiosensitiser in bladder cancer cell lines, and appears to target HR rather than NHEJ. As muscle-invasive bladder tumours have reduced Ku-DNA binding, PAN could be particularly useful as a radiosensitiser in bladder cancer.


Subject(s)
Antigens, Nuclear/physiology , DNA-Binding Proteins/physiology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Urinary Bladder Neoplasms/radiotherapy , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , DNA-Binding Proteins/analysis , Histones/analysis , Humans , Ku Autoantigen , MRE11 Homologue Protein , Panobinostat , Rad51 Recombinase/analysis , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...