Network


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

Hotspot


Dive into the research topics where Hirokazu Hirakawa is active.

Publication


Featured researches published by Hirokazu Hirakawa.


Radiotherapy and Oncology | 2012

Different effects of carbon ion beams and X-rays on clonogenic survival and DNA repair in human pancreatic cancer stem-like cells

Kazuhiko Oonishi; Xing Cui; Hirokazu Hirakawa; Akira Fujimori; Takehiko Kamijo; Shigeru Yamada; Osamu Yokosuka; Tadashi Kamada

PURPOSE The effects of a carbon ion beam and X-rays on human pancreatic cancer stem-like cells were examined from the point of view of clonogenic survival and DNA repair. MATERIALS AND METHODS Human pancreatic cancer stem-like cells were treated with and without carbon ion and X-ray irradiation, and then colony, spheroid and tumor formation assays as well as γH2AX foci formation assay were performed. RESULTS The relative biological effectiveness (RBE) values of a carbon ion beam relative to X-ray for the MIA PaCa-2 and BxPc-3 cells at the D10 values were 1.85-2.10. The ability for colony, spheroid formation, and tumorigenicity from cancer stem-like CD44(+)/CD24(+) cells is significantly higher than that from non-cancer stem-like CD44(-)/CD24(-)cells. FACS data showed that CD44(+)/CD24(+) cells were more highly enriched after X-rays compared to carbon ion irradiation at isoeffective doses. The RBE values for the carbon ion beam relative to X-ray at the D10 levels for CD44(+)/CD24(+) cells were 2.0-2.19. The number of γH2AX foci in CD44(-)/CD24(-) cells was higher than that of CD44(+)/CD24(+) cells after irradiation with either X-ray or carbon ion beam. The number of γH2AX foci in CD44(+)/CD24(+) cells was almost the same in the early time, but it persists significantly longer in carbon ion beam irradiated cells compared to X-rays. CONCLUSIONS Carbon ion beam has superior potential to kill pancreatic cancer stem cell-like cells, and prolonged induction of DNA damage might be one of the pivotal mechanisms of its high radiobiological effects compared to X-rays.


Brain & Development | 2014

Disruption of Aspm causes microcephaly with abnormal neuronal differentiation

Akira Fujimori; Kyoko Itoh; Shoko Goto; Hirokazu Hirakawa; Bing Wang; Toshiaki Kokubo; Seiji Kito; Satoshi Tsukamoto; Shinji Fushiki

AIMS A number of ASPM mutations have been detected in primary microcephaly patients. In order to evaluate the function of ASPM in brain development, we generated model animals of human autosomal recessive primary microcephaly-5 (MCPH5). METHODS In the Aspm knock-out mice, the exon 2-3 of the Aspm gene was encompassed by a pair of loxP signals so that cre-recombinase activity switched the allele from wild-type to null zygotes as frequently, as expected from the Mendelian inheritance. We precisely analyzed the brains of adults and fetuses using immunohistochemistry and morphometry. RESULTS The adult brains of the Aspm(-/-) mice were smaller, especially in the cerebrum. In the barrel field of the somatosensory cortex, layer I was significantly thicker, whereas layer VI was significantly thinner in Aspm(-/-) mice, compared with Aspm(+/+) mice. The total number of cells and the thickness of the cortical plate at embryonic day 16.5 was significantly decreased in Aspm(-/-) mice, compared with Aspm(+/+) mice. Furthermore, the expression of transcription factors, such as Tbr1 and Satb2, was significantly increased in the subplate of the Aspm(-/-) mice. CONCLUSIONS The results suggested that Aspm is essential to the proliferation and differentiation of neural stem/progenitor cells. The Aspm gene loss model provided a novel pathogenetic insight into acquired microcephaly, which can be caused by in utero exposure to both known and unknown teratogens.


Cancer Science | 2016

Nontoxic concentration of DNA-PK inhibitor NU7441 radio-sensitizes lung tumor cells with little effect on double strand break repair.

Shigeaki Sunada; Hideki Kanai; Younghyun Lee; Takeshi Yasuda; Hirokazu Hirakawa; Cuihua Liu; Akira Fujimori; Mitsuru Uesaka; Ryuichi Okayasu

High‐linear energy transfer (LET) heavy ions have been increasingly employed as a useful alternative to conventional photon radiotherapy. As recent studies suggested that high LET radiation mainly affects the nonhomologous end‐joining (NHEJ) pathway of DNA double strand break (DSB) repair, we further investigated this concept by evaluating the combined effect of an NHEJ inhibitor (NU7441) at a non‐toxic concentration and carbon ions. NU7441‐treated non‐small cell lung cancer (NSCLC) A549 and H1299 cells were irradiated with X‐rays and carbon ions (290 MeV/n, 50 keV/μm). Cell survival was measured by clonogenic assay. DNA DSB repair, cell cycle distribution, DNA fragmentation and cellular senescence induction were studied using a flow cytometer. Senescence‐associated protein p21 was detected by western blotting. In the present study, 0.3 μM of NU7441, nontoxic to both normal and tumor cells, caused a significant radio‐sensitization in tumor cells exposed to X‐rays and carbon ions. This concentration did not seem to cause inhibition of DNA DSB repair but induced a significant G2/M arrest, which was particularly emphasized in p53‐null H1299 cells treated with NU7441 and carbon ions. In addition, the combined treatment induced more DNA fragmentation and a higher degree of senescence in H1299 cells than in A549 cells, indicating that DNA‐PK inhibitor contributes to various modes of cell death in a p53‐dependent manner. In summary, NSCLC cells irradiated with carbon ions were radio‐sensitized by a low concentration of DNA‐PK inhibitor NU7441 through a strong G2/M cell cycle arrest. Our findings may contribute to further effective radiotherapy using heavy ions.


Journal of Radiation Research | 2014

High linear-energy-transfer radiation can overcome radioresistance of glioma stem-like cells to low linear-energy-transfer radiation

Yuki Hirota; Shin-ichiro Masunaga; Natsuko Kondo; Shinji Kawabata; Hirokazu Hirakawa; Hirohiko Yajima; Akira Fujimori; Koji Ono; Toshihiko Kuroiwa; Shin-Ichi Miyatake

Ionizing radiation is applied as the standard treatment for glioblastoma multiforme (GBM). However, radiotherapy remains merely palliative, not curative, because of the existence of glioma stem cells (GSCs), which are regarded as highly radioresistant to low linear-energy-transfer (LET) photons. Here we analyzed whether or not high-LET particles can overcome the radioresistance of GSCs. Glioma stem-like cells (GSLCs) were induced from the GBM cell line A172 in stem cell culture medium. The phenotypes of GSLCs and wild-type cells were confirmed using stem cell markers. These cells were irradiated with 60Co gamma rays or reactor neutron beams. Under neutron-beam irradiation, high-LET proton particles can be produced through elastic scattering or nitrogen capture reaction. Radiosensitivity was assessed by a colony-forming assay, and the DNA double-strand breaks (DSBs) were assessed by a histone gamma-H2AX focus detection assay. In stem cell culture medium, GSLCs could form neurosphere-like cells and express neural stem cell markers (Sox2 and Musashi) abundantly in comparison with their parental cells. GSLCs were significantly more radioresistant to gamma rays than their parental cells, but neutron beams overcame this resistance. There were significantly fewer gamma-H2AX foci in the A172 GSLCs 24 h after irradiation with gamma rays than in their parental cultured cells, while there was no apparent difference following neutron-beam irradiation. High-LET radiation can overcome the radioresistance of GSLCs by producing unrepairable DNA DSBs. High-LET radiation therapy might have the potential to overcome GBMs resistance to X-rays in a clinical setting.


Cancer Medicine | 2015

The combination of Hsp90 inhibitor 17AAG and heavy-ion irradiation provides effective tumor control in human lung cancer cells

Hirokazu Hirakawa; Hiroshi Fujisawa; Aya Masaoka; Miho Noguchi; Ryoichi Hirayama; Momoko Takahashi; Akira Fujimori; Ryuichi Okayasu

Hsp90 inhibitors have become well‐studied antitumor agents for their selective property against tumors versus normal cells. The combined treatment of Hsp90 inhibitor and conventional photon radiation also showed more effective tumor growth delay than radiation alone. However, little is known regarding the combined treatment of Hsp90 inhibitor and heavy‐ion irradiation. In this study, SQ5 human lung tumor cells were used in vitro for clonogenic cell survival and in vivo for tumor growth delay measurement using a mouse xenograft model after 17‐allylamino‐17‐demethoxygeldanamycin (17AAG) pretreatment and carbon ion irradiation. Repair of DNA double strand breaks (DSBs) was also assessed along with expressions of DSB repair‐related proteins. Cell cycle analysis after the combined treatment was also performed. The combined treatment of 17AAG and carbon ions revealed a promising treatment option in both in vitro and in vivo studies. One likely cause of this effectiveness was shown to be the inhibition of homologous recombination repair by 17AAG. The more intensified G2 cell cycle delay was also associated with the combined treatment when compared with carbon ion treatment alone. Our findings indicate that the combination of Hsp90 inhibition and heavy‐ion irradiation provides a new effective therapeutic alternative for treatment of solid tumors.


Molecular Cancer Therapeutics | 2017

TAS-116, a Novel Hsp90 Inhibitor, Selectively Enhances Radiosensitivity of Human Cancer Cells to X-rays and Carbon Ion Radiation.

Younghyun Lee; Shigeaki Sunada; Hirokazu Hirakawa; Akira Fujimori; Jac A. Nickoloff; Ryuichi Okayasu

Hsp90 inhibitors have been investigated as cancer therapeutics in monotherapy and to augment radiotherapy; however, serious adverse effects of early-generation Hsp90 inhibitors limited their development. TAS-116 is a novel Hsp90 inhibitor with lower adverse effects than other Hsp90 inhibitors, and here, we investigated the radiosensitizing effects of TAS-116 in low linear energy transfer (LET) X-ray and high LET carbon ion–irradiated human cancer cells and mouse tumor xenografts. TAS-116 decreased cell survival of both X-ray and carbon ion–irradiated human cancer cell lines (HeLa and H1299 cells), and similar to other Hsp90 inhibitors, it did not affect radiosensitivity of noncancerous human fibroblasts. TAS-116 increased the number of radiation-induced γ-H2AX foci and delayed the repair of DNA double-strand breaks (DSB). TAS-116 reduced the expression of proteins that mediate repair of DSBs by homologous recombination (RAD51) and nonhomologous end joining (Ku, DNA-PKcs), and suppressed formation of RAD51 foci and phosphorylation/activation of DNA-PKcs. TAS-116 also decreased expression of the cdc25 cell-cycle progression marker, markedly increasing G2–M arrest. Combined treatment of mouse tumor xenografts with carbon ions and TAS-116 showed promising delay in tumor growth compared with either individual treatment. These results demonstrate that TAS-116 radiosensitizes human cancer cells to both X-rays and carbon ions by inhibiting the two major DSB repair pathways, and these effects were accompanied by marked cell-cycle arrest. The promising results of combination TAS-116 + carbon ion radiotherapy of tumor xenografts justify further exploration of TAS-116 as an adjunct to radiotherapy using low or high LET radiation. Mol Cancer Ther; 16(1); 16–24. ©2016 AACR.


Radiotherapy and Oncology | 2016

The purine scaffold Hsp90 inhibitor PU-H71 sensitizes cancer cells to heavy ion radiation by inhibiting DNA repair by homologous recombination and non-homologous end joining

Younghyun Lee; Huizi Keiko Li; Aya Masaoka; Shigeaki Sunada; Hirokazu Hirakawa; Akira Fujimori; Jac A. Nickoloff; Ryuichi Okayasu

BACKGROUND AND PURPOSE PU-H71 is a purine-scaffold Hsp90 inhibitor developed to overcome limitations of conventional Hsp90 inhibitors. This study was designed to investigate the combined effect of PU-H71 and heavy ion irradiation on human tumor and normal cells. MATERIALS AND METHODS The effects of PU-H71 were determined by monitoring cell survival by colony formation, and DNA double-strand break (DSB) repair by γ-H2AX foci and immuno-blotting DSB repair proteins. The mode of cell death was evaluated by sub-G1 DNA content (as an indicator for apoptosis), and mitotic catastrophe. RESULTS PU-H71 enhanced heavy ion irradiation-induced cell death in three human cancer cell lines, but the drug did not radiosensitize normal human fibroblasts. In irradiated tumor cells, PU-H71 increased the persistence of γ-H2AX foci, and it reduced RAD51 foci and phosphorylated DNA-PKcs, key DSB repair proteins involved in homologous recombination (HR) and non-homologous end joining (NHEJ). In some tumor cell lines, PU-H71 altered the sub-G1 cell fraction and mitotic catastrophe following carbon ion irradiation. CONCLUSION Our results demonstrate that PU-H71 sensitizes human cancer cells to heavy ion irradiation by inhibiting both HR and NHEJ DSB repair pathways. PU-H71 holds promise as a radiosensitizer for enhancing the efficacy of heavy ion radiotherapy.


International Journal of Radiation Biology | 2014

Carbon ion beam is more effective to induce cell death in sphere-type A172 human glioblastoma cells compared with X-rays

Momoko Takahashi; Hirokazu Hirakawa; Hirohiko Yajima; Nakako Izumi-Nakajima; Ryuichi Okayasu; Akira Fujimori

Abstract Purpose: To obtain human glioblastoma cells A172 expressing stem cell-related protein and comparison of radiosensitivity in these cells with X-rays and carbon beam. Methods: Human monolayer-type A172 glioblastoma cells were maintained in normal medium with 10% bovine serum. In order to obtain sphere-type A172 cells the medium was replaced with serum-free medium supplemented with growth factors. Both types of A172 cells were irradiated with either X-rays or carbon ion beams and their radiosensitivity was evaluated. Results: Serum-free medium induced expression of stem cell-related proteins in A172 cells along with the neurosphere-like appearance. These sphere-type cells were found resistant to both X-rays and carbon ion beams. Phosphorylation of histone H2A family member X persisted for a longer period in the cells exposed to carbon ion beams than in those exposed to X-rays and it disappeared quicker in the sphere type than in the monolayer type. Relative radioresistance of the sphere type cells was smaller for carbon ion beams than for X-rays. Conclusions: We demonstrated that glioblastoma A172 cells with induced stem cell-related proteins turned resistant to irradiation. Accelerated heavy ion particles may have advantage over X-rays in overcoming the tumor resistance due to cell stemness.


Oncology Letters | 2015

Hyperthermia-induced radiosensitization in CHO wild-type, NHEJ repair mutant and HR repair mutant following proton and carbon-ion exposure

Junko Maeda; Yoshihiro Fujii; Hiroshi Fujisawa; Hirokazu Hirakawa; Ian M. Cartwright; Mitsuru Uesaka; Hisashi Kitamura; Akira Fujimori; Takamitsu A. Kato

The DNA repair mechanisms involved in hyperthermia-induced radiosensitization with proton and carbon ion radiation exposure were investigated in the present study. In a previous study, Chinese hamster ovary (CHO) cells were exposed to low linear energy transfer (LET) photon radiation. These cells can be sensitized by hyperthermia as a result of inhibition of homologous recombination (HR) repair. The present study used wild-type, non-homologous end joining (NHEJ) and HR repair-deficient CHO cells to define the contributions of each repair pathway to cellular lethality following hyperthermia-induced hadron radiation sensitization. The cells were exposed to ionizing radiation, followed by hyperthermia treatment (42.5°C for 1 h). Hyperthermia-induced radiosensitization was determined by the colony formation assay and thermal enhancement ratio. HR repair-deficient cells exhibited no hyper-sensitization to X-rays, protons, or low and high LET carbon ions when combined with hyperthermia. Wild-type and NHEJ repair-deficient cells exhibited significant hyperthermia-induced sensitization to low LET photon and hadron radiation. Hyperthermia-induced sensitization to high LET carbon-ion radiation was less than at low LET radiation. Relative biological effectiveness (RBE) between radiation alone and radiation combined with hyperthermia cell groups was not significantly different in any of the cell lines, with the exception of wild-type cells exposed to high LET radiation, which exhibited a lower RBE in the combined group. The present study investigated additional cell lines to confirm the lower RBE observed in DNA repair-deficient cell lines. These findings suggested that hyperthermia-induced hyper-sensitization to hadron radiation is also dependent on inhibition of HR repair, as was observed with photon radiation in a previous study.


Journal of Radiation Research | 2014

Hsp90 inhibitor is a good candidate for effective combination therapy with carbon ions

Ryuichi Okayasu; Hirokazu Hirakawa; Aya Masaoka; Miho Noguchi; Ryoichi Hirayama; Huizi Li; Yoshitaka Matsumoto; Akira Fujimori

Purpose/Background: To further improve the effectiveness of heavy ion radio-therapy, we studied the biology of a combined treatment using Hsp 90 inhibitor and carbon ion irradiation. We have previously reported that an Hsp90 inhibitor 17AAG can be an effective radio-sensitizer with X-rays for certain human tumor cells, while normal cells were not sensitized by this drug [ 1]. The underlying mechanism for this was demonstrated to be inhibition of DNA double-strand break (DSB) repair by 17AAG; particularly, homologous recombination repair (HRR) pathway was shown to be affected by this agent. In vivo mouse xenograft study also indicated a better tumor control with the combined treatment when compared with X-ray treatment alone. Materials and Methods: Cell lines used were SQ5 human lung cancer cells and HFL III normal human fibroblasts as control. For irradiation, X-rays and 290 MeV/n carbon ions were used at 50–70 keV/µm LET setting. As Hsp90 inhibitors, 17AAG and PU-H71 were used for pre-irradiation treatment for 24 h. Colony formation assay was used for radiation sensitivity studies. Repair of DNA DSBs was measured by constant field gel electrophoresis, and the appearance/disappearance of Rad51 foci was analyzed for HRR efficiency. For IRB-approved mouse xenograft study, BALB/c nu/nu mice were used to implant SQ5 cells for local irradiation. Results/Discussion: SQ5 tumor cells were better controlled with the combination of 17 AAG and carbon ion irradiation in vitro and in vivo xenograft model when compared with carbon irradiation alone. The cause of this radio-sensitizing effect seems to come from inhibition of repair of DNA DSBs by 17AAG as in X-rays. Likewise, HRR pathway was affected by addition of 17AAG in carbon irradiated tumor cells. The effect of 17AAG pre-treatment is shown below where the appearance of an HRR key protein Rad51 was significantly delayed after carbon ion irradiation in the drug-treated samples when compared with samples with carbon alone (Fig. 1). We also started to investigate PU-H71 with carbon ion irradiation in vitro and in vivo. PU-H71 alone is currently under phase I clinical trial. Our data indicate that PU-H71 pre-treatment also showed a significant radio-sensitization in SQ5 human lung tumor cells exposed to carbon ions as well as to X-rays. As shown with 17AAG, normal human cells were not significantly affected with this drug. PU-H71 also seems to affect repair of radiation-induced DSBs. Further mechanism studies and in vivo experiments are currently underway. Conclusion: Hsp90 inhibitor would be a good candidate for the effective combined treatment with carbon ion radio-therapy. Fig. 1. Post-irradiation Rad51 foci appearance kinetics in SQ5 cells irradiated with 2 Gy carbon ions compared with the combined treatment of 17AAG and carbon radiation (2 Gy).

Collaboration


Dive into the Hirokazu Hirakawa's collaboration.

Top Co-Authors

Avatar

Akira Fujimori

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Ryuichi Okayasu

Nuclear Information and Resource Service

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aya Masaoka

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Miho Noguchi

Japan Atomic Energy Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hirohiko Yajima

National Institute of Radiological Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge