Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Norio Suzuki is active.

Publication


Featured researches published by Norio Suzuki.


Oncogene | 2001

Variations in Prkdc encoding the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) and susceptibility to radiation-induced apoptosis and lymphomagenesis

Nobuko Mori; Yoshihisa Matsumoto; Masaaki Okumoto; Norio Suzuki; Jyoji Yamate

DNA double-strand breaks (DSBs) induced by ionizing radiation enforce cells to die, if unrepaired; while if misrepaired, DSBs may cause malignant transformation. The DSB repair system predominant in mammals requires DNA-dependent protein kinase (DNA-PK). Previously, we identified the apoptosis susceptibility gene Radiation-induced apoptosis 1 (Rapop1) on mouse chromosome 16. The STS/A (STS) allele at Rapop1 leads to decreased sensitivity to apoptosis in the BALB/cHeA (BALB/c) background. In the present study, we established Rapop1 congenic strains C.S-R1 and C.S-R1L, which contain the STS genome in a 0.45 cM interval critical for Rapop1 in common in the BALB/c background. Within the segment critical for Rapop1, Prkdc encoding the catalytic subunit of DNA-PK (DNA-PKcs) was assigned. Two variations T6,418C and G11,530A, which induce amino acid substitutions C2,140R downstream from the putative leucine zipper motif and V3,844M near the kinase domain, respectively, were found between BALB/c and STS for Prkdc. The majority of inbred strains such as C57BL/6J carried the STS allele at Prkdc; a few strains including 129/SvJ and C.B17 carried the BALB/c allele. DNA-PK activity as well as DNA-PKcs expression was profoundly diminished in BALB/c and 129/SvJ mice as compared with C57BL/6 and C.S-R1 mice. In the crosses (C.S-R1 x BALB/c)F1 x 129/SvJ and (C.S-R1 x BALB/c)F1 x C.B17, enhanced apoptosis occurred in the absence of the wild-type allele at Prkdc. C.S-R1 and C.S-R1L were both less sensitive to radiation lymphomagenesis than BALB/c. Our study provides strong evidence for Prkdc as a candidate for Rapop1 and a susceptibility gene for radiation lymphomagenesis as well.


FEBS Letters | 2000

Cleavage and phosphorylation of XRCC4 protein induced by X-irradiation

Yoshihisa Matsumoto; Norio Suzuki; Naoki Namba; Noriko Umeda; Xue-Jun Ma; Akinori Morita; Masanori Tomita; Atsushi Enomoto; Shinobu Serizawa; Kazuya Hirano; Kazuo Sakai; Hideyo Yasuda; Yoshio Hosoi

We report the p35 and p60 forms of XRCC4 protein, appearing in human leukemia MOLT‐4 or U937 cells following X‐irradiation or hyperthermia. p35 appeared in conjunction with the cleavage of DNA‐dependent protein kinase catalytic subunit (DNA‐PKcs) and the fragmentation of internucleosomal DNA, and was suppressed by Ac‐DEVD‐CHO. p35 was also produced in vitro by treating MOLT‐4 cell lysate with recombinant caspases, suggesting that p35 was a caspase‐cleaved fragment of XRCC4 in apoptotic cell death. p60 was sensitive to treatment with phosphatase or wortmannin and was undetectable in M059J cells deficient in DNA‐PKcs. However, p60 was found in ataxia‐telangiectasia cells after irradiation. These results indicated p60 as a phosphorylated form of XRCC4, requiring DNA‐PKcs but not ataxia‐telangiectasia mutated (ATM).


Cancer Letters | 2000

Involvement of SAPK/JNK pathway in X-ray-induced rapid cell death of human T-cell leukemia cell line MOLT-4

Atsushi Enomoto; Norio Suzuki; Kazuya Hirano; Yoshihisa Matsumoto; Akinori Morita; Kazuo Sakai; Hideki Koyama

We found that SAPK/JNK was phosphorylated during X-ray-induced rapid cell death of MOLT-4 cells and that acid Sphingomyelinase inhibitor D609 suppressed the rapid cell death as well as phosphorylation of SAPK/JNK. Also C2-ceramide caused phosphorylation of SAPK/JNK, followed by rapid cell death. Further we isolated X-ray-resistant radiation-hybrid clones from MOLT-4 and 50 Gy irradiated mouse FM3A cells by repeated selections with 3 Gy irradiation. One of them named Rh-1a was found resistant to X-ray- as well as C2-ceramide-induced rapid cell death. Rh-1a cells had mouse DNA but no increase in either mouse or human Bcl-2 determined by Western blotting. Accumulation of p53 after X-irradiation was similarly observed in both parental MOLT-4 and Rh-1a cells. However, contrasting to prolonged and prominent phosphorylated status of SAPK/JNK in MOLT-4 cells, Rh-1a cells exhibited short transient increase and FM3A cells showed no increase of phosphorylated status SAPK/JNK after X-irradiation. Therefore, SAPK/JNK activation is considered important in X-ray-induced rapid cell death or apoptosis of MOLT-4 cells.


International Journal of Radiation Oncology Biology Physics | 2001

Expression of genes involved in repair of DNA double-strand breaks in normal and tumor tissues

Koh-ichi Sakata; Yoshihisa Matsumoto; Hiroshi Tauchi; Masaaki Satoh; Atushi Oouchi; Hisayasu Nagakura; Kazumitsu Koito; Yoshio Hosoi; Norio Suzuki; Kenshi Komatsu; Masato Hareyama

BACKGROUNDnDNA double-strand breaks (DSB) are the major lethal lesions induced by ionizing radiation. The capability for DNA DSB repair is crucial for inherent radiosensitivity of tumor and normal cells. DNA-PKcs, Ku 70, Ku 85, Xrcc4, and Nbs1 play a critical role in DNA DSB repair.nnnMETHODSnWe immunohistochemically investigated the expression of DNA-PKcs, Ku 70, Ku85, Xrcc4, and Nbs1 in 134 specimens from various normal and tumor tissues with different radiosensitivity.nnnRESULTS AND CONCLUSIONnImmunopositivity to Ku70, Ku85, DNA-PKcs, Xrcc4, and Nbs1 was found in all tumor tissues examined. The staining for Ku70, Ku85, and DNA-PKcs was nuclear; but, for Xrcc4 and Nbs1, it was nuclear and cytoplasmic. There were no apparent differences in the expression of these five proteins among cancerous tissues and the corresponding normal tissues. No apparent differences in nuclear staining intensity were detected in the expression of these five proteins among tumor tissues with different radiosensitivity, although non-Hodgkins lymphoma (B or T cell) tended to show a lower expression than the others. The stromal cells generally expressed these five proteins at much lower frequency than either tumor or epithelial cells in both tumor and normal tissues.


Cell Death & Differentiation | 2006

Sodium orthovanadate suppresses DNA damage- induced caspase activation and apoptosis by inactivating p53

Akinori Morita; Jin Zhu; Norio Suzuki; Atsushi Enomoto; Yoshihisa Matsumoto; Masanori Tomita; Takahiko Suzuki; Kuni Ohtomo; Yoshio Hosoi

We previously reported that p42/SETβ is a substrate for caspase-7 in irradiated MOLT-4 cells, and that treating the cells with sodium orthovanadate (vanadate) inhibits p42/SETβs caspase-mediated cleavage. Here, we initially found that the inhibitory effect of vanadate was due to the suppression of caspase activation but not of caspase activity. Further investigations revealed that vanadate suppressed upstream of apoptotic events, such as the loss of mitochondrial membrane potential, the conformational change of Bax, and p53 transactivation, although the accumulation, total phosphorylation, and phosphorylation of six individual sites of p53 were not affected. Importantly, vanadate suppressed p53-dependent apoptosis, but not p53-independent apoptosis. Finally, gel-shift and chromatin immunoprecipitation assays conclusively demonstrated that vanadate inhibits the DNA-binding activity of p53. Vanadate is conventionally used as an inhibitor of protein tyrosine phosphatases (PTPs); however, we recommend that the influence of vanadate not only on PTPs but also on p53 be considered before using it.


Biochemical and Biophysical Research Communications | 2003

Caspase-mediated cleavage of JNK during stress-induced apoptosis

Atsushi Enomoto; Norio Suzuki; Akinori Morita; Michihiko Ito; Chang Qing Liu; Yoshihisa Matsumoto; Katsuji Yoshioka; Tadayoshi Shiba; Yoshio Hosoi

The c-Jun N-terminal kinases (JNKs) are a subfamily of the mitogen-activated protein kinases (MAPKs). The JNKs are encoded by three separate genes (jnk1, jnk2, and jnk3), which are spliced alternatively to create 10 JNK isoforms that are either p46 or p54 in size. In this study, we found that the p52 form of JNK emerged in human leukemia MOLT-4 or U937 cells following X-irradiation or heat treatment. The accumulation of p52 coincided with the reduction of p54 JNK. On the other hand, the amounts of p46 JNK did not change by X-irradiation. Induction of the p52 form of JNK also paralleled the appearance of the active form of caspase-3 and was suppressed by a caspase-specific inhibitor, Ac-DEVD-CHO, but not by Ac-YVAD-CHO. In vitro cleavage assays indicated that recombinant human JNK1beta2 and JNK2beta2 were cleaved by caspase-3, and that the mutation of aspartic acid at position 413 of JNK1beta2 or 410 of JNK2beta2 to alanine abolished the cleavage. Altogether, our results demonstrated that p54 JNKs, at least JNK1beta2 and JNK2beta2, were new selective targets of caspases in JNK splicing variants, and suggested that the p52 form could serve as a marker of apoptosis.


International Journal of Radiation Biology | 2002

Roles of DNA-dependent protein kinase and ATM in cell-cycle-dependent radiation sensitivity in human cells.

Masayuki Yoshida; Yoshio Hosoi; H. Miyachi; N. Ishii; Yoshihisa Matsumoto; Atsushi Enomoto; Keiichi Nakagawa; S. Yamada; Norio Suzuki; T. Ono

Purpose : The roles of DNA-dependent protein kinase (DNA-PK) and ATM in the cell-cycle-dependent radiosensitivity in human cells were investigated. Methods and materials : A DNA-PK activity-deficient human glioblastoma cell line M059J, ataxia telangiectasia cell lines AT3BISV and AT5BIVA, and control cell lines were used. Wortmannin inhibited DNA-PK and ATM activities. Cells were synchronized by hydroxyurea. Progression through the cell cycle was analysed by flow cytometry. Results : M059J exhibited hyper-radiosensitivity throughout the cell cycle, with extreme hyper-radiosensitivity in G to early S-phase compared with the control cell line M059K. AT3BISV and AT5BIVA exhibited hyper-radiosensitivity throughout the cell cycle but showed a similar pattern of cell-cycle-dependent radiosensitivity to that observed in LM217 or HeLa cells. In AT3BISV and AT5BIVA, radiosensitization by wortmannin was observed throughout the cell cycle and was most prominent in G 1 to early S-phase. Wortmannin did not sensitize M059J to ionizing radiation in any cell-cycle phase. DNA-PK activities were not different throughout the cell cycle. Conclusion : The results suggest that (1) non-homologous endjoining plays a dominant role in G 1 to early S-phase and a minor role in late S to G 2 -phase in repairing DNA double-strand breaks, (2) the role of ATM in repairing double-strand breaks may be almost cell-cycle-independent and (3) the dominant role of non-homologous end-joining during G 1 to early S-phase is not due to cell-cycle-dependent fluctuations in DNA-PK activity.


Radiation Research | 2003

Wortmannin-Enhanced X-Ray-Induced Apoptosis of Human T-Cell Leukemia MOLT-4 Cells Possibly through the JNK/SAPK Pathway

Masanori Tomita; Norio Suzuki; Yoshihisa Matsumoto; Atsushi Enomoto; Hong Lan Yin; Yoshio Hosoi; Kazuya Hirano; Kazuo Sakai

Abstract Tomita, M., Suzuki, N., Matsumoto, Y., Enomoto, A., Yin, H. L., Hosoi, Y., Hirano, K. and Sakai, K. Wortmannin-Enhanced X-Ray-Induced Apoptosis of Human T-Cell Leukemia MOLT-4 Cells Possibly through the JNK/SAPK Pathway. Radiat. Res. 160, 467–477 (2003). We demonstrated that enhancement of X-ray-induced apoptosis/rapid cell death by wortmannin accompanied by increased activation of JNK/SAPK in human leukemia MOLT-4 cells. Rapid cell death/apoptosis was determined either by the dye exclusion test or by the appearance of Annexin V-positive cells and cleaved PARP fragments. Enhancement was observed only at higher concentrations of wortmannin, i.e. 1 μM or more. At these high concentrations, both DNA-PK and ATM were inhibited. X-ray-induced phosphorylation of Ser 15 of p53/TP53, accumulation of both p53/TP53 and p21/WAF1/CDKN1A, and phosphorylation of XRCC4 were all suppressed. The enhancement of apoptosis/rapid cell death by wortmannin was prevented by addition of caspase inhibitors, Z-VAD-FMK or Ac-DEVD-CHO, or by transfection and overexpression of mouse Bcl2, which is known as an anti-apoptosis protein. The requirement for a high concentration of wortmannin, i.e. 1 μM or more, indicates that inhibition of both DNA-PK and ATM was necessary for the enhanced apoptosis/rapid cell death. Phosphorylation of AKT/PKB was completely suppressed at a much lower concentration, i.e. 0.1 μM wortmannin, where no enhancement of X-ray-induced apoptosis/rapid cell death was observed. On the other hand, X-ray-induced phosphorylation of JNK and its kinase activity as well as apoptosis/rapid cell death were all significantly enhanced only at high concentrations of wortmannin, i.e. 1 μM or more. Furthermore, the extent of enhancement of both JNK phosphorylation and of apoptosis/rapid cell death by wortmannin was less in Rh1a cells, which are ceramide- and radiation-resistant variant cells compared to the parental MOLT-4 cells. Therefore, activation of the JNK pathway was considered important for the enhancement of X-ray-induced apoptosis/rapid cell death of MOLT-4 cells by wortmannin, because of the requirement for a higher concentration of wortmannin than that required for inhibition of AKT phosphorylation. The suppression of the AKT-dependent pathway by wortmannin may have some underlying role in activating the JNK pathway toward the enhancement of cell death in the current system.


Japanese Journal of Cancer Research | 2000

Increased Expression after X‐Irradiation of MUC1 in Cultured Human Colon Carcinoma HT‐29 Cells

Yun Kang; Kazuya Hirano; Norio Suzuki; Atsushi Enomoto; Akinori Morita; Tatsuro Irimura; Kazuo Sakai

The effect of X‐irradiation on production of MUC1 was studied with human colon carcinoma HT‐29 cells. As evaluated by immunocytochemical staining, the percentages of MUC1‐positive cells in cells at 4 days after 6 Gy irradiation and in unirradiated control cells were 52±3.5% (n=6) and 26±2.8% (n=6), respectively. Flow‐cytometric analysis of living cells showed that MUC1 began to rise from day 1, reaching a plateau by day 4 after 6 Gy irradiation. Western blot analysis with monoclonal antibody MY.1E12 against glycosylated MUC1 (mature form) showed dose‐dependent increases of two bands (500 and 390 kDa) corresponding to two polymorphic MUC1 alleles. Premature forms of MUC1 (350 and 240 kDa) were detectable with monoclonal antibody HMFG‐2 only in irradiated cells, suggesting that new core protein synthesis had been induced. The transcriptional activity of the MUC1 gene was analyzed in terms of transient expression of MUC1‐CAT reporter plasmids containing 5′‐flanking sequences of the MUC1 gene fused to the bacterial chloramphenicol acetyltransferase (CAT) gene. The results of CAT assay indicate that enhanced expression of MUC1 in irradiated HT‐29 cells was due to upregulation of MUC1 transcription, and required the upstream promoter.


International Journal of Radiation Biology | 2003

Difference in the heat sensitivity of DNA‐dependent protein kinase activity among mouse, hamster and human cells

Noriko Umeda; Yoshihisa Matsumoto; Hong-Lan Yin; Masanori Tomita; Atsushi Enomoto; Akinori Morita; T. Mizukoshi; Kazuo Sakai; Yoshio Hosoi; Norio Suzuki

Purpose: To examine the heat sensitivity of DNA‐dependent protein kinase (DNA‐PK) activity in a variety of cultured mouse, hamster and human cell lines. Materials and methods: Eight cell lines, which have been routinely used in our laboratory, were examined. Cells were heated at 44.0±0.05°C and DNA‐PK activity was measured by a DNA‐pull‐down assay followed by gel‐electrophoresis. Cellular sensitivity to hyperthermia and/or X‐ray was evaluated by a colony formation assay. Results: In mouse FSA1233 and FM3A cells, DNA‐PK activity dropped to 15–16% of unheated control after 20u2009min of heating. In Chinese hamster V79 and CHO‐K1 cells, kinase activity did not change appreciably after 20u2009min treatment but decreased to 60–70 and 22–23% after 40 or 60u2009min treatment, respectively. However, even after 180u2009min treatment, DNA‐PK activity remained almost intact in human MOLT‐4, MKN45 and A7 cells, and decreased only slightly in U937 cells. Hyperthermic radiosensitization was seen even in human cells but, as a trend, it was small compared with rodent cells. Conclusions: The heat sensitivity of DNA‐PK was clearly different among mouse, hamster and human cells. The results suggested a possibility that the role of DNA‐PK inactivation in hyperthermic radiosensitization might be variable, depending on cells, and would reinforce the warning that the direct extrapolation of data from rodent cells might lead to overestimation of the effectiveness of hyperthermia on human cancer.

Collaboration


Dive into the Norio Suzuki's collaboration.

Top Co-Authors

Avatar

Yoshihisa Matsumoto

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Koh-ichi Sakata

Sapporo Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge