Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37345118

ABSTRACT

CERS6 is associated with metastasis and poor prognosis in non-small cell lung cancer (NSCLC) patients through d18:1/C16:0 ceramide (C16 ceramide)-mediated cell migration, though the detailed mechanism has not been elucidated. In the present study, examinations including co-immunoprecipitation, liquid chromatography, and tandem mass spectrometry analysis were performed to identify a novel binding partner of CERS6. Among the examined candidates, LASP1 was a top-ranked binding partner, with the LIM domain possibly required for direct interaction. In accord with those findings, CERS6 and LASP1 were found to co-localize on lamellipodia in several lung cancer cell lines. Furthermore, silencing of CERS6 and/or LASP1 significantly suppressed cell migration and lamellipodia formation, whereas ectopic addition of C16 ceramide partially rescued those phenotypes. Both LASP1 and CERS6 showed co-immunoprecipitation with actin, with those interactions markedly reduced when the LASP1-CERS6 complex was abolished. Based on these findings, it is proposed that LASP1-CERS6 interaction promotes cancer cell migration.

2.
Acta Histochem Cytochem ; 55(5): 129-148, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36405554

ABSTRACT

The enzyme-labeled antigen method is an immunohistochemical technique detecting plasma cells producing specific antibodies in tissue sections. The probe is an antigen labeled with an enzyme or biotin. This immunohistochemical technique is appliable to frozen sections of paraformaldehyde (PFA)-fixed tissues, but it has been difficult to apply it to formalin-fixed, paraffin-embedded (FFPE) sections. In the current study, factors inactivating the antibody reactivity during the process of preparing FFPE sections were investigated. Lymph nodes of rats immunized with horseradish peroxidase (HRP) or a mixture of keyhole limpet hemocyanin/ovalbumin/bovine serum albumin were employed as experimental models. Plasma cells producing specific antibodies, visualized with HRP (as an antigen with enzymatic activity) or biotinylated proteins in 4% PFA-fixed frozen sections, significantly decreased in unbuffered 10% formalin-fixed frozen sections. The positive cells were further decreased by paraffin embedding following formalin fixation. In paraffin-embedded sections fixed in precipitating fixatives such as ethanol and acetone and those prepared with the AMeX method, the antigen-binding reactivity of antibodies was preserved. Fixation in periodate-lysine-paraformaldehyde and Zamboni solution also kept the antigen-binding reactivity in paraffin to some extent. In conclusion, formalin fixation and paraffin embedding were major causes inactivating antibodies. Precipitating fixatives could retain the antigen-binding reactivity of antibodies in paraffin-embedded sections.

3.
J Clin Med ; 11(18)2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36143113

ABSTRACT

Alanine-serine-cysteine transporter 2 (ASCT2) has been associated with increased levels of metabolism in various malignant tumors. However, its biological significance in the proliferation of prostate cancer (PCa) cells remains under investigation. We used the cBioPortal database to assess the effect of ASCT2 expression on the oncological outcomes of 108 PCa patients. To evaluate the function of ASCT2 in castration-sensitive PCa (CSPC) and castration-resistant PCa (CRPC), LNCaP cells and the ARV7-positive PCa cell line, 22Rv1, were assessed using cell proliferation assays and Western blot analyses. The ASCT2 expression level was associated with biochemical recurrence-free survival after prostatectomy in patients with a Gleason score ≥ 7. In vitro experiments indicated that the growth of LNCaP cells after combination therapy of ASCT2 siRNA and enzalutamide treatment was significantly reduced, compared to that following treatment with enzalutamide alone or ASCT2 siRNA transfection alone (p < 0.01, 0.01, respectively). After ASCT2 inhibition by siRNA transfection, the growth of 22Rv1 cells was significantly suppressed as compared with negative control siRNA via downregulation of ARV7 both in fetal bovine serum and androgen-deprivation conditions (p < 0.01, 0.01, respectively). We demonstrated that ASCT2 inhibition significantly reduced the proliferation rates of both CSPC and CRPC cells in vitro.

4.
Pediatr Surg Int ; 38(8): 1157-1163, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35699751

ABSTRACT

PURPOSE: We previously reported that polyphyllin D, a main component of the traditional Chinese medicinal herb Paris polyphylla, exhibited anticancer effects in vitro against human neuroblastoma cells. The aims of this investigation was to examine the presence or absence of in vivo anti-metastasis effects of polyphyllin D were to establish a liver metastasis model of neuroblastoma and to evaluate the anti-metastasis effects of polyphyllin D. METHODS: Subcutaneous and intraperitoneal tumors, and metastasis models were established in immune-deficient BALB/c nude and BALB/c Rag-2/Jak3 double-deficient (BRJ) mice using the human neuroblastoma cell lines IMR-32, LA-N-2, or NB-69. For evaluating polyphyllin D activity, we used a mouse model of liver metastasis with the IMR-32 cells line injected through the tail vein. We analyzed the livers number and area of liver tumors in of the phosphate buffer solution- and polyphyllin D-treated groups. RESULTS: Liver metastasis and intraperitoneal dissemination models were successfully established in immune-deficient BRJ mice using the three human neuroblastoma cell lines. In the liver metastasis, the model of IMR-32 cells, we found that polyphyllin D suppressed both the number and total area of metastatic foci the average number of metastatic foci, average focus areas, and number of cleaved caspase-3-positive cells were significantly lower in the polyphyllin D group (p = 0.016, 0.020, 0.043, respectively). CONCLUSIONS: We developed a mouse models of neuroblastoma metastasis and demonstrated for the first time that polyphyllin D has an antitumor effect on neuroblastoma liver metastases.


Subject(s)
Diosgenin , Liver Neoplasms , Neuroblastoma , Animals , Apoptosis , Cell Line, Tumor , Diosgenin/analogs & derivatives , Diosgenin/pharmacology , Liver Neoplasms/drug therapy , Mice , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Saponins
5.
Cancer Sci ; 112(7): 2770-2780, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33934437

ABSTRACT

Ceramide synthase 6 (CERS6) promotes lung cancer metastasis by stimulating cancer cell migration. To examine the underlying mechanisms, we performed luciferase analysis of the CERS6 promoter region and identified the Y-box as a cis-acting element. As a parallel analysis of database records for 149 non-small-cell lung cancer (NSCLC) cancer patients, we screened for trans-acting factors with an expression level showing a correlation with CERS6 expression. Among the candidates noted, silencing of either CCAAT enhancer-binding protein γ (CEBPγ) or Y-box binding protein 1 (YBX1) reduced the CERS6 expression level. Following knockdown, CEBPγ and YBX1 were found to be independently associated with reductions in ceramide-dependent lamellipodia formation as well as migration activity, while only CEBPγ may have induced CERS6 expression through specific binding to the Y-box. The mRNA expression levels of CERS6, CEBPγ, and YBX1 were positively correlated with adenocarcinoma invasiveness. YBX1 expression was observed in all 20 examined clinical lung cancer specimens, while 6 of those showed a staining pattern similar to that of CERS6. The present findings suggest promotion of lung cancer migration by possible involvement of the transcription factors CEBPγ and YBX1.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Pseudopodia , Sphingosine N-Acyltransferase/metabolism , Y-Box-Binding Protein 1/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Neoplasm Invasiveness , Promoter Regions, Genetic , Pseudopodia/genetics , RNA, Messenger/metabolism , Sphingosine N-Acyltransferase/genetics , Transcriptional Activation , Up-Regulation , Y-Box-Binding Protein 1/genetics , rac1 GTP-Binding Protein
6.
Prostate Int ; 9(1): 18-24, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33912510

ABSTRACT

BACKGROUND: Disease progression in castrate-resistant prostate cancer (PCa) is most commonly driven by the reactivation of androgen receptor (AR) signaling and involves AR splice variants including ARV7. MATERIALS AND METHODS: We used the ARV7-positive PCa cell line, 22Rv1, to study the relationship of the PCa marker α-methylacyl-CoA racemase (AMACR), AR, and ARV7 in PCa. RESULTS: Docetaxel addition but not AMACR inhibition decreased the proliferation of 22Rv1 cells. The combination of AMACR inhibition and docetaxel treatment resulted in a maximum reduction of cell proliferation. The Western blotting analysis revealed that both AR and ARV7 expression were significantly decreased with the use of charcoal-stripped serum following AMACR inhibition and docetaxel treatment. AMACR inhibition and docetaxel treatment in the charcoal-stripped serum condition reduced the proliferation of 22Rv1, possibly via the downregulation of the heat shock protein 27. CONCLUSION: Using cell proliferation and Western blot analysis, we demonstrated that AMACR inhibition and docetaxel treatment, under androgen deprivation conditions, significantly reduced the proliferation of ARV7 positive cancer cells and decreased the levels of AR and ARV7 expression, possibly via downregulation of heat shock protein 27.

7.
J Cell Mol Med ; 24(20): 11949-11959, 2020 10.
Article in English | MEDLINE | ID: mdl-32902157

ABSTRACT

Sphingolipids constitute a class of bio-reactive molecules that transmit signals and exhibit a variety of physical properties in various cell types, though their functions in cancer pathogenesis have yet to be elucidated. Analyses of gene expression profiles of clinical specimens and a panel of cell lines revealed that the ceramide synthase gene CERS6 was overexpressed in non-small-cell lung cancer (NSCLC) tissues, while elevated expression was shown to be associated with poor prognosis and lymph node metastasis. NSCLC profile and in vitro luciferase analysis results suggested that CERS6 overexpression is promoted, at least in part, by reduced miR-101 expression. Under a reduced CERS6 expression condition, the ceramide profile became altered, which was determined to be associated with decreased cell migration and invasion activities in vitro. Furthermore, CERS6 knockdown suppressed RAC1-positive lamellipodia/ruffling formation and attenuated lung metastasis efficiency in mice, while forced expression of CERS6 resulted in an opposite phenotype in examined cell lines. Based on these findings, we consider that ceramide synthesis by CERS6 has important roles in lung cancer migration and metastasis.


Subject(s)
Cell Movement , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Ceramides/metabolism , Humans , Male , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Neoplasm Metastasis , Pseudopodia/metabolism , Treatment Outcome
8.
J Radiat Res ; 60(1): 69-79, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30476166

ABSTRACT

Photons, such as X- or γ-rays, induce DNA damage (distributed throughout the nucleus) as a result of low-density energy deposition. In contrast, particle irradiation with high linear energy transfer (LET) deposits high-density energy along the particle track. High-LET heavy-ion irradiation generates a greater number and more complex critical chromosomal aberrations, such as dicentrics and translocations, compared with X-ray or γ irradiation. In addition, the formation of >1000 bp deletions, which is rarely observed after X-ray irradiation, has been identified following high-LET heavy-ion irradiation. Previously, these chromosomal aberrations have been thought to be the result of misrepair of complex DNA lesions, defined as DNA damage through DNA double-strand breaks (DSBs) and single-strand breaks as well as base damage within 1-2 helical turns (<3-4 nm). However, because the scale of complex DNA lesions is less than a few nanometers, the large-scale chromosomal aberrations at a micrometer level cannot be simply explained by complex DNA lesions. Recently, we have demonstrated the existence of clustered DSBs along the particle track through the use of super-resolution microscopy. Furthermore, we have visualized high-level and frequent formation of DSBs at the chromosomal boundary following high-LET heavy-ion irradiation. In this review, we summarize the latest findings regarding the hallmarks of DNA damage structure and the repair pathway following heavy-ion irradiation. Furthermore, we discuss the mechanism through which high-LET heavy-ion irradiation may induce dicentrics, translocations and large deletions.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Heavy Ions , Chromosomes/radiation effects , Histones/metabolism , Humans , Translational Research, Biomedical
9.
Hum Immunol ; 79(8): 627-631, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29859207

ABSTRACT

Programmed cell death-1 (PD-1) and its ligand (programmed death-ligand 1, PD-L1) are key factors that regulate a cytotoxic immune reaction. Anti-PD-1 therapy provides significant clinical benefits for patients with cancer, even those with advanced-stage cancer. We have recently demonstrated that DNA damage signaling from DNA double-strand breaks (DSBs) promotes PD-L1 upregulation in cancer cells. In the present study, we aimed to investigate PD-L1 expression in primary normal human dermal fibroblasts (NHDFs) in response to DSBs. We demonstrated that PD-L1 expression in NHDFs is not upregulated after ionizing radiation (IR). In addition, interferon (IFN) regulatory factor 1 (IRF1) and signal transducer and activator of transcription 1 (STAT1) phosphorylation do not respond in NHDFs after IR. In contrast, IFNγ treatment upregulates PD-L1 and IRF1 expressions and STAT1 phosphorylation. The nonresponsiveness was also observed after treatment with other DNA-damaging agents, such as camptothecin and etoposide. Treatment with a histone deacetylase inhibitor (HDACi), which causes chromatin relaxation and restores gene silencing, upregulates PD-L1 without exogenous DNA damage; however, IR-dependent upregulation is not observed in NHDFs treated with HDACi. Taken together, our data suggest that DNA-damage signaling is insufficient for upregulating PD-L1 in NHDFs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/metabolism , DNA Damage/immunology , Dermis/pathology , Fibroblasts/physiology , Immunotherapy/methods , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cells, Cultured , Etoposide/pharmacology , Gene Expression Regulation , Histone Deacetylases/metabolism , Humans , Interferon Regulatory Factor-1/metabolism , Primary Cell Culture , Radiation, Ionizing , STAT1 Transcription Factor/metabolism
10.
Nat Commun ; 8(1): 1751, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29170499

ABSTRACT

Accumulating evidence suggests that exogenous cellular stress induces PD-L1 upregulation in cancer. A DNA double-strand break (DSB) is the most critical type of genotoxic stress, but the involvement of DSB repair in PD-L1 expression has not been investigated. Here we show that PD-L1 expression in cancer cells is upregulated in response to DSBs. This upregulation requires ATM/ATR/Chk1 kinases. Using an siRNA library targeting DSB repair genes, we discover that BRCA2 depletion enhances Chk1-dependent PD-L1 upregulation after X-rays or PARP inhibition. In addition, we show that Ku70/80 depletion substantially enhances PD-L1 upregulation after X-rays. The upregulation by Ku80 depletion requires Chk1 activation following DNA end-resection by Exonuclease 1. DSBs activate STAT1 and STAT3 signalling, and IRF1 is required for DSB-dependent PD-L1 upregulation. Thus, our findings reveal the involvement of DSB repair in PD-L1 expression and provide mechanistic insight into how PD-L1 expression is regulated after DSBs.


Subject(s)
B7-H1 Antigen/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Neoplasms/genetics , Neoplasms/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , B7-H1 Antigen/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Humans
11.
Oncol Rep ; 38(2): 693-702, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28677817

ABSTRACT

Immunotherapy is expected to be promising as a next generation cancer therapy. Immunoreceptors are often activated constitutively in cancer cells, however, such levels of ligand expression are not effectively recognized by the native immune system due to tumor microenvironmental adaptation. Studies have demonstrated that natural-killer group 2, member D (NKG2D), a major activating immunoreceptor, responds to DNA damage. The upregulation of major histocompatibility complex class I-related chain A and B (MICA/B) (members of NKG2D ligands) expression after DNA damage is associated with NK cell-mediated killing of cancer cells. However, the regulation of DNA damage-induced MICA/B expression has not been fully elucidated in the context of the types of cancer cell lines. In the present study, we found that MICA/B expression varied between cancer cell lines after DNA damage. Screening in terms of chromatin remodeling identified that inhibitors related to chromatin relaxation via post-translational modification on histone H3K9, i.e. HDAC, Suv39 or G9a inhibition, restored DNA damage-dependent MICA/B expression in insensitive cells. In addition, we revealed that the restored MICA/B expression was dependent on ATR as well as E2F1, a transcription factor. We further revealed that low­dose treatment of an HDAC inhibitor was sufficient to restore MICA/B expression in insensitive cells. Finally, we demonstrated that HDAC inhibition restored DNA damage­dependent cytotoxic NK activity against insensitive cells. Thus, the present study revealed that DNA damage­dependent MICA/B expression in insensitive cancer cells can be restored by chromatin relaxation via the HDAC/Suv39/G9a pathway. Collectively, manipulation of chromatin status by therapeutic cancer drugs may potentiate the antitumor effect by enhancing immune activation following radiotherapy and DNA damage-associated chemotherapy.


Subject(s)
Histocompatibility Antigens Class I/genetics , Histone Deacetylases/genetics , Neoplasms/immunology , Ataxia Telangiectasia Mutated Proteins/genetics , Chromatin Assembly and Disassembly/genetics , Cytotoxicity, Immunologic/genetics , DNA Damage/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens/genetics , Histocompatibility Antigens Class I/immunology , Histone Deacetylases/immunology , Histone-Lysine N-Methyltransferase/genetics , Humans , Killer Cells, Natural/immunology , Methyltransferases/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/genetics , Neoplasms/pathology , Repressor Proteins/genetics , Tumor Microenvironment/immunology
12.
Sci Rep ; 7: 41812, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28155885

ABSTRACT

Chromosome rearrangement is clinically and physiologically important because it can produce oncogenic fusion genes. Chromosome rearrangement requires DNA double-strand breaks (DSBs) at two genomic locations and misrejoining between the DSBs. Before DSB misrejoining, two DSB-containing chromatin regions move and pair with each other; however, the molecular mechanism underlying this process is largely unknown. We performed a spatiotemporal analysis of ionizing radiation-induced foci of p53-binding protein 1 (53BP1), a marker for DSB-containing chromatin. We found that some 53BP1 foci were paired, indicating that the two damaged chromatin regions neighboured one another. We searched for factors regulating the foci pairing and found that the number of paired foci increased when Ku80, DNA-PKcs, or ATM was absent. In contrast, 53BP1 depletion reduced the number of paired foci and dicentric chromosomes-an interchromosomal rearrangement. Foci were paired more frequently in heterochromatin than in euchromatin in control cells. Additionally, the reduced foci pairing in 53BP1-depleted cells was rescued by concomitant depletion of a heterochromatin building factor such as Krüppel-associated box-associated protein 1 or chromodomain helicase DNA-binding protein 3. These findings indicate that pairing between DSB-containing chromatin regions was suppressed by Ku80, DNA-PKcs, and ATM, and this pairing was promoted by 53BP1 through chromatin relaxation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Ku Autoantigen/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Biomarkers , Chromatin/radiation effects , Chromosome Aberrations , DNA Breaks, Double-Stranded/radiation effects , DNA-Binding Proteins/metabolism , Fibroblasts , Fluorescent Antibody Technique , Humans , Protein Binding , Radiation, Ionizing , Signal Transduction
13.
Cell Rep ; 18(2): 520-532, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076794

ABSTRACT

BRCA1 promotes homologous recombination (HR) by activating DNA-end resection. By contrast, 53BP1 forms a barrier that inhibits DNA-end resection. Here, we show that BRCA1 promotes DNA-end resection by relieving the 53BP1-dependent barrier. We show that 53BP1 is phosphorylated by ATM in S/G2 phase, promoting RIF1 recruitment, which inhibits resection. 53BP1 is promptly dephosphorylated and RIF1 released, despite remaining unrepaired DNA double-strand breaks (DSBs). When resection is impaired by CtIP/MRE11 endonuclease inhibition, 53BP1 phosphorylation and RIF1 are sustained due to ongoing ATM signaling. BRCA1 depletion also sustains 53BP1 phosphorylation and RIF1 recruitment. We identify the phosphatase PP4C as having a major role in 53BP1 dephosphorylation and RIF1 release. BRCA1 or PP4C depletion impairs 53BP1 repositioning, EXO1 recruitment, and HR progression. 53BP1 or RIF1 depletion restores resection, RAD51 loading, and HR in PP4C-depleted cells. Our findings suggest that BRCA1 promotes PP4C-dependent 53BP1 dephosphorylation and RIF1 release, directing repair toward HR.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Homologous Recombination , Signal Transduction , Tumor Suppressor p53-Binding Protein 1/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair Enzymes/metabolism , Endodeoxyribonucleases , Exodeoxyribonucleases/metabolism , G2 Phase , Humans , MRE11 Homologue Protein/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , S Phase , Telomere-Binding Proteins/metabolism
14.
Sci Rep ; 7: 40588, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091564

ABSTRACT

In cancer therapy today, carbon ion radiotherapy is used mainly as monotherapy, whereas cisplatin is used concomitantly with X-ray radiotherapy. The effectiveness of concomitant carbon ions and cisplatin is unclear. To obtain the information on the mechanisms potentially shared between carbon ions or X-rays and cisplatin, we assessed the correlation of sensitivity to the single treatments. In 20 human cancer cell lines, sensitivity to X-rays strongly correlated with sensitivity to cisplatin, indicating the presence of potentially shared target mechanisms. Interestingly, the correlation of sensitivity to carbon ions and cisplatin was much weaker than that of sensitivity to X-rays and cisplatin, indicating the presence of potentially different target mechanisms between carbon ions and cisplatin. Assessment of clonogenic cell death by 4',6-diamidino-2-phenylindole dihydrochloride staining showed that mitotic catastrophe was more efficiently induced by carbon ions than by the same physical dose of X-rays, while apoptosis and senescence were not. These data indicate that the correlation of sensitivity to carbon ions and cisplatin is weaker than that of sensitivity to X-rays and cisplatin, which are helpful as biological basis to understand the potentially shared mechanism among these treatments. Further investigation is mandatory to elucidate the clinical efficacy of carbon ions and cisplatin combination.


Subject(s)
Cisplatin/pharmacology , Heavy Ion Radiotherapy , Mitosis/drug effects , Mitosis/radiation effects , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Clone Cells , Humans , X-Rays
15.
Oncotarget ; 8(65): 109370-109381, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29312614

ABSTRACT

DNA double-strand breaks (DSBs) induced by ionising radiation are considered the major cause of genotoxic mutations and cell death. While DSBs are dispersed throughout chromatin after X-rays or γ-irradiation, multiple types of DNA damage including DSBs, single-strand breaks and base damage can be generated within 1-2 helical DNA turns, defined as a complex DNA lesion, after high Linear Energy Transfer (LET) particle irradiation. In addition to the formation of complex DNA lesions, recent evidence suggests that multiple DSBs can be closely generated along the tracks of high LET particle irradiation. Herein, by using three dimensional (3D)-structured illumination microscopy, we identified the formation of 3D widespread γH2AX foci after high LET carbon-ion irradiation. The large γH2AX foci in G2-phase cells encompassed multiple foci of replication protein A (RPA), a marker of DSBs undergoing resection during homologous recombination. Furthermore, we demonstrated by 3D analysis that the distance between two individual RPA foci within γH2AX foci was approximately 700 nm. Together, our findings suggest that high LET heavy-ion particles induce clustered DSB formation on a scale of approximately 1 µm3. These closely localised DSBs are considered to be a risk for the formation of chromosomal rearrangement after heavy-ion irradiation.

16.
Genes Chromosomes Cancer ; 55(8): 650-60, 2016 08.
Article in English | MEDLINE | ID: mdl-27113385

ABSTRACT

Chromosomal translocations arise from misrejoining of DNA double strand breaks (DSBs) between loci located on two chromosomes. One current model suggests that spatial proximity of potential chromosomal translocation partners influences translocation probability. Ionizing radiation (IR) is a potent inducer of translocations. Accumulating evidence demonstrates that particle irradiation more frequently causes translocations compared with X-ray irradiation. This observation has led to the hypothesis that the high frequency of translocations after particle irradiation may be due to the formation of DSBs at chromosome boundaries along the particle track, because such DSBs can be misrejoined between distinct chromosomes. In this study, we simultaneously visualized the site of IR-induced DSBs and chromosome position by combining Immunofluorescence and fluorescence in situ hybridization. Importantly, the frequency of γH2AX foci at the chromosome boundary of chromosome 1 after carbon-ion irradiation was >4-fold higher than that after X-ray irradiation. This observation is consistent with the idea that particle irradiation generates DSBs at the boundaries of two chromosomes along the track. Further, we showed that resolution of γH2AX foci at chromosome boundaries is prevented by inhibition of DNA-PKcs activity, indicating that the DSB repair is NHEJ-dependent. Finally, we found that γH2AX foci at chromosome boundaries after carbon-ion irradiation contain DSBs undergoing DNA-end resection, which promotes repair utilizing microhomology mediated end-joining during translocation. Taken together, our study suggests that the frequency of DSB formation at chromosome boundaries is associated with the incidence of chromosomal translocations, supporting the notion that the spatial proximity between breaks is an important factor in translocation formation. © 2016 Wiley Periodicals, Inc.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , Histones/genetics , Translocation, Genetic/radiation effects , Carbon Radioisotopes , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Fibroblasts/radiation effects , Humans , In Situ Hybridization, Fluorescence , Radiation, Ionizing , X-Rays
17.
Sci Rep ; 6: 22275, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26925533

ABSTRACT

Carbon ion radiotherapy shows great potential as a cure for X-ray-resistant tumors. Basic research suggests that the strong cell-killing effect induced by carbon ions is based on their ability to cause complex DNA double-strand breaks (DSBs). However, evidence supporting the formation of complex DSBs in actual patients is lacking. Here, we used advanced high-resolution microscopy with deconvolution to show that complex DSBs are formed in a human tumor clinically treated with carbon ion radiotherapy, but not in a tumor treated with X-ray radiotherapy. Furthermore, analysis using a physics model suggested that the complexity of radiotherapy-induced DSBs is related to linear energy transfer, which is much higher for carbon ion beams than for X-rays. Visualization of complex DSBs in clinical specimens will help us to understand the anti-tumor effects of carbon ion radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA/ultrastructure , Heavy Ion Radiotherapy , Uterine Cervical Neoplasms/radiotherapy , Adult , Aged , Biopsy , Cell Death/radiation effects , DNA Breaks, Double-Stranded/radiation effects , Female , Humans , Linear Energy Transfer , Microscopy , Tumor Burden/radiation effects , Tumor Suppressor p53-Binding Protein 1/immunology , Uterine Cervical Neoplasms/ultrastructure , X-Ray Therapy
18.
Mutat Res ; 779: 16-23, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26117423

ABSTRACT

Monoubiquitination of proliferating cell nuclear antigen (PCNA) is a critical regulator of post replication repair (PRR). The depletion of BAF180, a unique subunit of the PBAF chromatin remodeling complex in human cells results in reduced PCNA ubiquitination leading to less efficient fork progression following DNA damage, but little is known about the mechanism. Here, we report that the expression of exogenous BAF180 in cells promotes PCNA ubiquitination during S-phase after UV irradiation and it persists for many hours. No correlation was observed between the protein level of ubiquitin-specific protease 1 (USP1) and ubiquitinated PCNA in BAF180 expressing cells. Analysis of cells expressing BAF180 deletion mutants showed that the bromo-adjacent homology (BAH) domains are responsible for this effect. Surprisingly, a deletion construct encoding only the BAH domain region is able to increase the level of ubiquitinated PCNA, even though it is unable to be assembled into the PBAF complex. These results suggest that the ATPase-dependent chromatin remodeling activity of PBAF is not necessary, but instead the BAH domains are sufficient to promote PCNA ubiquitination.


Subject(s)
Arabidopsis Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Proliferating Cell Nuclear Antigen/biosynthesis , Transcription Factors/biosynthesis , Ubiquitin-Specific Proteases/biosynthesis , Ubiquitination/radiation effects , Adenosine Triphosphatases/metabolism , Arabidopsis Proteins/genetics , Cell Line , Chromatin Assembly and Disassembly/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA Replication/radiation effects , DNA-Binding Proteins , Gene Expression Regulation/radiation effects , Humans , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/genetics , Protein Structure, Tertiary , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics , Ultraviolet Rays
19.
Anal Biochem ; 443(2): 211-3, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24012794

ABSTRACT

We demonstrate that firefly luciferase is a good reporter in Escherichia coli for transcription dynamics in response to the environment. E. coli strains, carrying a fusion of the promoter of the ycgZ gene and the coding region of the luciferase gene, showed transient bioluminescence on receiving blue light. This response was compromised in mutants lacking known regulators in manners consistent with each regulator's function. We also show that relA, a gene encoding a (p)ppGpp synthetase, affects ycgZ dynamics when nullified. Moreover, two unstable luciferase variants showed improved response dynamics and should be useful to study quick changes of gene expression.


Subject(s)
Escherichia coli/genetics , Fireflies/enzymology , Genes, Bacterial/genetics , Genes, Reporter , Luciferases, Firefly/genetics , Transcriptional Activation , Animals , Gene Expression Regulation, Bacterial , Light
20.
Nucleic Acids Res ; 40(15): 7393-403, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22638582

ABSTRACT

In eukaryotic cells, replication past damaged sites in DNA is regulated by the ubiquitination of proliferating cell nuclear antigen (PCNA). Little is known about how this process is affected by chromatin structure. There are two isoforms of the Remodels the Structure of Chromatin (RSC) remodelling complex in yeast. We show that deletion of RSC2 results in a dramatic reduction in the level of PCNA ubiquitination after DNA-damaging treatments, whereas no such effect was observed after deletion of RSC1. Similarly, depletion of the BAF180 component of the corresponding PBAF (Polybromo BRG1 (Brahma-Related Gene 1) Associated Factor) complex in human cells led to a similar reduction in PCNA ubiquitination. Remarkably, we found that depletion of BAF180 resulted after UV-irradiation, in a reduction not only of ubiquitinated PCNA but also of chromatin-associated unmodified PCNA and Rad18 (the E3 ligase that ubiquitinates PCNA). This was accompanied by a modest decrease in fork progression. We propose a model to account for these findings that postulates an involvement of PBAF in repriming of replication downstream from replication forks blocked at sites of DNA damage. In support of this model, chromatin immunoprecipitation data show that the RSC complex in yeast is present in the vicinity of the replication forks, and by extrapolation, this is also likely to be the case for the PBAF complex in human cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle , Cell Line , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/physiology , Gene Deletion , Humans , Nuclear Proteins/antagonists & inhibitors , Proliferating Cell Nuclear Antigen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , Ubiquitin-Protein Ligases , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...