Network


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

Hotspot


Dive into the research topics where Kazuaki Matsuoka is active.

Publication


Featured researches published by Kazuaki Matsuoka.


Molecular Cancer Therapeutics | 2015

Trifluridine induces p53-dependent sustained G2 phase arrest with its massive misincorporation into DNA and few DNA strand breaks

Kazuaki Matsuoka; Makoto Iimori; Shinichiro Niimi; Hiroshi Tsukihara; Sugiko Watanabe; Shinichi Kiyonari; Mamoru Kiniwa; Koji Ando; Eriko Tokunaga; Hiroshi Saeki; Eiji Oki; Yoshihiko Maehara; Hiroyuki Kitao

Trifluridine (FTD) is a key component of the novel oral antitumor drug TAS-102, which consists of FTD and a thymidine phosphorylase inhibitor. Like 5-fluoro-2′-deoxyuridine (FdUrd), a deoxynucleoside form of 5-fluorouracil metabolite, FTD is sequentially phosphorylated and not only inhibits thymidylate synthase activity, but is also incorporated into DNA. Although TAS-102 was effective for the treatment of refractory metastatic colorectal cancer in clinical trials, the mechanism of FTD-induced cytotoxicity is not completely understood. Here, we show that FTD as well as FdUrd induce transient phosphorylation of Chk1 at Ser345, and that this is followed by accumulation of p53 and p21 proteins in p53-proficient human cancer cell lines. In particular, FTD induced p53-dependent sustained arrest at G2 phase, which was associated with a proteasome-dependent decrease in the Cyclin B1 protein level and the suppression of CCNB1 and CDK1 gene expression. In addition, a p53-dependent increase in p21 protein was associated with an FTD-induced decrease in Cyclin B1 protein. Although numerous ssDNA and dsDNA breaks were induced by FdUrd, few DNA strand breaks were detected in FTD-treated HCT-116 cells despite massive FTD misincorporation into genomic DNA, suggesting that the antiproliferative effect of FTD is not due to the induction of DNA strand breaks. These distinctive effects of FTD provide insights into the cellular mechanism underlying its antitumor effect and may explain the clinical efficacy of TAS-102. Mol Cancer Ther; 14(4); 1004–13. ©2015 AACR.


Nature Communications | 2016

Phosphorylation of EB2 by Aurora B and CDK1 ensures mitotic progression and genome stability

Makoto Iimori; Sugiko Watanabe; Shinichi Kiyonari; Kazuaki Matsuoka; Ryo Sakasai; Hiroshi Saeki; Eiji Oki; Hiroyuki Kitao; Yoshihiko Maehara

Temporal regulation of microtubule dynamics is essential for proper progression of mitosis and control of microtubule plus-end tracking proteins by phosphorylation is an essential component of this regulation. Here we show that Aurora B and CDK1 phosphorylate microtubule end-binding protein 2 (EB2) at multiple sites within the amino terminus and a cluster of serine/threonine residues in the linker connecting the calponin homology and end-binding homology domains. EB2 phosphorylation, which is strictly associated with mitotic entry and progression, reduces the binding affinity of EB2 for microtubules. Expression of non-phosphorylatable EB2 induces stable kinetochore microtubule dynamics and delays formation of bipolar metaphase plates in a microtubule binding-dependent manner, and leads to aneuploidy even in unperturbed mitosis. We propose that Aurora B and CDK1 temporally regulate the binding affinity of EB2 for microtubules, thereby ensuring kinetochore microtubule dynamics, proper mitotic progression and genome stability.


DNA Repair | 2012

ATR–Chk1 signaling pathway and homologous recombinational repair protect cells from 5-fluorouracil cytotoxicity

Yoshihiko Fujinaka; Kazuaki Matsuoka; Makoto Iimori; Munkhbold Tuul; Ryo Sakasai; Keiji Yoshinaga; Hiroshi Saeki; Masaru Morita; Yoshihiro Kakeji; David A. Gillespie; Ken Yamamoto; Minoru Takata; Hiroyuki Kitao; Yoshihiko Maehara

5-Fluorouracil (5-FU) has long been a mainstay antimetabolite chemotherapeutic drug for the treatment of major solid tumors, particularly colorectal cancer. 5-FU is processed intracellularly to yield active metabolites that compromise RNA and DNA metabolism. However, the mechanisms responsible for its cytotoxicity are not fully understood. From the phenotypic analysis of mutant chicken B lymphoma DT40 cells, we found that homologous recombinational repair (HRR), involving Rad54 and BRCA2, and the ATR-Chk1 signaling pathway, involving Rad9 and Rad17, significantly contribute to 5-FU tolerance. 5-FU induced γH2AX nuclear foci, which were colocalized with the key HRR factor Rad51, but not with DNA double-strand breaks (DSBs), in a dose-dependent manner as cells accumulated in the S phase. Inhibition of Chk1 kinase by UCN-01 increased 5-FU-induced γH2AX and enhanced 5-FU cytotoxicity not only in wild-type cells but also in Rad54- or BRCA2-deficient cells, suggesting that HRR and Chk1 kinase have non-overlapping roles in 5-FU tolerance. 5-FU-induced Chk1 phosphorylation was significantly impaired in Rad9- or Rad17-deficient cells, and severe γH2AX nuclear foci and DSBs were formed, which was followed by apoptosis. Finally, inhibition of Chk1 kinase by UCN-01 increased 5-FU-induced γH2AX nuclear foci and enhanced 5-FU cytotoxicity in Rad9- or Rad17-deficient cells. These results suggest that Rad9- and Rad17-independent activation of the ATR-Chk1 signaling pathway also significantly contributes to 5-FU tolerance.


PLOS ONE | 2013

Rad9, Rad17, TopBP1 and Claspin Play Essential Roles in Heat-Induced Activation of ATR Kinase and Heat Tolerance

Munkhbold Tuul; Hiroyuki Kitao; Makoto Iimori; Kazuaki Matsuoka; Shinichi Kiyonari; Hiroshi Saeki; Eiji Oki; Masaru Morita; Yoshihiko Maehara

Hyperthermia is widely used to treat patients with cancer, especially in combination with other treatments such as radiation therapy. Heat treatment per se activates DNA damage responses mediated by the ATR-Chk1 and ATM-Chk2 pathways but it is not fully understood how these DNA damage responses are activated and affect heat tolerance. By performing a genetic analysis of human HeLa cells and chicken B lymphoma DT40 cells, we found that heat-induced Chk1 Ser345 phosphorylation by ATR was largely dependent on Rad9, Rad17, TopBP1 and Claspin. Activation of the ATR-Chk1 pathway by heat, however, was not associated with FancD2 monoubiquitination or RPA32 phosphorylation, which are known as downstream events of ATR kinase activation when replication forks are stalled. Downregulation of ATR, Rad9, Rad17, TopBP1 or Claspin drastically reduced clonogenic cell viability upon hyperthermia, while gene knockout or inhibition of ATM kinase reduced clonogenic viability only modestly. Suppression of the ATR-Chk1 pathway activation enhanced heat-induced phosphorylation of Chk2 Thr68 and simultaneous inhibition of ATR and ATM kinases rendered severe heat cytotoxicity. These data indicate that essential factors for activation of the ATR-Chk1 pathway at stalled replication forks are also required for heat-induced activation of ATR kinase, which predominantly contributes to heat tolerance in a non-overlapping manner with ATM kinase.


Scientific Reports | 2016

The antibodies against 5-bromo-2′-deoxyuridine specifically recognize trifluridine incorporated into DNA

Hiroyuki Kitao; Yosuke Morodomi; Shinichiro Niimi; Mamoru Kiniwa; Kazuhiko Shigeno; Kazuaki Matsuoka; Yuki Kataoka; Makoto Iimori; Eriko Tokunaga; Hiroshi Saeki; Eiji Oki; Yoshihiko Maehara

Trifluridine (FTD) is a key component of the novel oral antitumor drug TAS-102 (also named TFTD), which consists of FTD and a thymidine phosphorylase inhibitor. FTD is supposed to exert its cytotoxicity via massive misincorporation into DNA, but the underlying mechanism of FTD incorporation into DNA and its correlation with cytotoxicity are not fully understood. The present study shows that several antibodies against 5-bromo-2′-deoxyuridine (BrdU) specifically cross-react with FTD, either anchored to bovine serum albumin or incorporated into DNA. These antibodies are useful for several biological applications, such as fluorescence-activated cell sorting, fluorescent immunostaining and immunogold detection for electron microscopy. These techniques confirmed that FTD is mainly incorporated in the nucleus during S phase in a concentration-dependent manner. In addition, FTD was also detected by immunohistochemical staining in paraffin-embedded HCT-116 xenograft tumors after intraperitoneal administration of FTD. Intriguingly, FTD was hardly detected in surrounding matrices, which consisted of fibroblasts with marginal expression of the nucleoside transporter genes SLC29A1 and SLC29A2. Thus, applications using anti-BrdU antibodies will provide powerful tools to unveil the underlying mechanism of FTD action and to predict or evaluate the efficacy and adverse effects of TAS-102 clinically.


Molecular Cancer Therapeutics | 2015

The 1,2-Diaminocyclohexane Carrier Ligand in Oxaliplatin Induces p53-Dependent Transcriptional Repression of Factors Involved in Thymidylate Biosynthesis.

Shinichi Kiyonari; Makoto Iimori; Kazuaki Matsuoka; Sugiko Watanabe; Tomomi Morikawa-Ichinose; Daisuke Miura; Shinichiro Niimi; Hiroshi Saeki; Eriko Tokunaga; Eiji Oki; Masaru Morita; Kenji Kadomatsu; Yoshihiko Maehara; Hiroyuki Kitao

Platinum-based chemotherapeutic drugs are widely used as components of combination chemotherapy in the treatment of cancer. One such drug, oxaliplatin, exerts a synergistic effect against advanced colorectal cancer in combination with 5-fluorouracil (5-FU) and leucovorin. In the p53-proficient colorectal cancer cell line HCT116, oxaliplatin represses the expression of deoxyuridine triphosphatase (dUTPase), a ubiquitous pyrophosphatase that catalyzes the hydrolysis of dUTP to dUMP and inhibits dUTP-mediated cytotoxicity. However, the underlying mechanism of this activity has not been completely elucidated, and it remains unclear whether factors other than downregulation of dUTPase contribute to the synergistic effect of 5-FU and oxaliplatin. In this study, we found that oxaliplatin and dachplatin, platinum-based drugs containing the 1,2-diaminocyclohexane (DACH) carrier ligand, repressed the expression of nuclear isoform of dUTPase (DUT-N), whereas cisplatin and carboplatin did not. Oxaliplatin induced early p53 accumulation, upregulation of primary miR-34a transcript expression, and subsequent downregulation of E2F3 and E2F1. Nutlin-3a, which activates p53 nongenotoxically, had similar effects. Introduction of miR-34a mimic also repressed E2F1 and DUT-N expression, indicating that this miRNA plays a causative role. In addition to DUT-N, oxaliplatin repressed, in a p53-dependent manner, the expression of genes encoding enzymes involved in thymidylate biosynthesis. Consequently, oxaliplatin significantly decreased the level of dTTP in the dNTP pool in a p53-dependent manner. These data indicate that the DACH carrier ligand in oxaliplatin triggers signaling via the p53–miR-34a–E2F axis, leading to transcriptional regulation that ultimately results in accumulation of dUTP and reduced dTTP biosynthesis, potentially enhancing 5-FU cytotoxicity. Mol Cancer Ther; 14(10); 2332–42. ©2015 AACR.


Oncotarget | 2018

Trifluridine/tipiracil overcomes the resistance of human gastric 5-fluorouracil-refractory cells with high thymidylate synthase expression

Kazuaki Matsuoka; Fumio Nakagawa; Takashi Kobunai; Teiji Takechi

Trifluridine/tipiracil (FTD/TPI or TFTD, also known as TAS-102) is a combination of the antineoplastic thymidine analog, FTD, and thymidine phosphorylase inhibitor, TPI (molar ratio 1:0.5). FTD/TPI was approved in Japan, the United States, and the European Union for the treatment of unresectable advanced or recurrent colorectal cancer. We evaluated the in vitro and in vivo efficacy and mechanisms of action of FTD and FTD/TPI against 5-fluorouracil (5-FU)-resistant MKN45/5FU, MKN74/5FU, and KATOIII/5FU human gastric cancer cells overexpressing thymidylate synthase (TS) and their respective parent cell lines. MKN45/5FU and KATOIII/5FU cells were not cross-resistant to FTD, whereas MKN45/5FU cells were 3.7-fold more resistant than the parental cells in vitro. FTD was also incorporated into genomic DNA in a concentration-dependent manner in 5-FU-resistant and parental cells. Additionally, deoxyuridine monophosphate levels in MKN45/5FU cells after 24-h FTD treatment were 3.0-fold higher than those in parental cells, and FTD treatment for 72 h induced G2/M arrest in MKN45/5FU cells, unlike the S phase arrest in MKN45 cells. Thus, TS-overexpressing MKN45/5FU cells, but not MKN74/5FU and KATOIII/5FU cells, showed partial cross-resistance to FTD. However, FTD/TPI (administered orally twice a day) exhibited antitumor activity to the same extent in MKN45 and MKN45/5FU xenograft mouse models, overcoming in vitro cross-resistance to FTD. DNA incorporation rather than TS inhibition seems to be the main action of FTD under these in vivo conditions. Thus, FTD/TPI is a promising chemotherapeutic agent against gastric cancers recurring following 5-FU therapy.


Genes to Cells | 2012

CtIP‐ and ATR‐dependent FANCJ phosphorylation in response to DNA strand breaks mediated by DNA replication

Ryo Sakasai; Akiko Sakai; Makoto Iimori; Shinichi Kiyonari; Kazuaki Matsuoka; Yoshihiro Kakeji; Hiroyuki Kitao; Yoshihiko Maehara

FANCJ, also called BACH1/BRIP1, is a 5′‐3′ DEAH helicase, whose mutations are known as a risk factor for Fanconi anemia and also breast and ovarian cancer. FANCJ is thought to contribute to DNA double‐strand break (DSB) repair and S‐phase checkpoint through binding to multiple partner proteins, such as BRCA1 and TopBP1, but its molecular regulation remains unclear. We focused on DNA damage‐induced phosphorylation of FANCJ and found that reagents that cause DSB or replication fork stalling induce FANCJ hyperphosphorylation. In particular, camptothecin (CPT) induced rapid and efficient FANCJ hyperphosphorylation that was largely dependent on TopBP1 and ATM‐Rad3 related (ATR) kinase. Furthermore, DNA end resection that exposes single‐strand DNA at the DSB site was required for hyperphosphorylation. Interestingly, upon CPT treatment, a dramatic increase in the FANCJ–TopBP1 complex was observed, and this increase was not alleviated even when ATR‐dependent hyperphosphorylation was suppressed. These results suggest that FANCJ function may be modulated by hyperphosphorylation in a DNA end resection‐ and ATR‐dependent manner and by FANCJ–TopBP1 complex formation in response to replication‐coupled DSBs.


International Journal of Molecular Sciences | 2018

Effective Sequential Combined Chemotherapy with Trifluridine/Tipiracil and Regorafenib in Human Colorectal Cancer Cells

Kazuaki Matsuoka; Fumio Nakagawa; Nozomu Tanaka; Hiroyuki Okabe; Kenichi Matsuo; Teiji Takechi

Salvage chemotherapy for refractory metastatic colorectal cancer using trifluridine/tipiracil (FTD/TPI) and regorafenib has shown survival benefits. We evaluated the antitumor effects of FTD or FTD/TPI combined with regorafenib in vitro and in vivo. SW620, HCT 116, and HT-29 human colorectal cancer cell lines were treated with FTD and regorafenib simultaneously and sequentially. Cell death, incorporation of FTD into DNA, and molecules related to FTD and regorafenib-associated cell death were investigated. The antitumor effects of FTD combined with regorafenib in SW620 and COLO205 xenografts were also evaluated. Cell death was greater after sequential treatment with FTD followed by regorafenib in SW620 cells, but not in HCT 116 and HT-29 cells, than after treatment with FTD alone, which was attributable to thymidylate synthase reduction with the induction of apoptosis. In contrast, simultaneous and sequential exposure to regorafenib followed by FTD, but not FTD alone, attenuated the cell death effect. Furthermore, combined FTD/TPI treatment followed by regorafenib had greater antitumor activity than either monotherapy in SW620 and COLO205 xenograft models. Treatment results following regorafenib administration subsequent to FTD or FTD/TPI suggest that sequential therapy with FTD/TPI prior to regorafenib may be effective in a clinical setting.


Cancer Research | 2016

Abstract 5175: Mutational and copy number profiling of cancer-related genes in 26 human tumor xenografts and their correlations with antitumor drug sensitivities

Takashi Kobunai; Kenta Tsunekuni; Kazuaki Matsuoka; Hiroshi Tsukihara; Teiji Takechi

Background: Tumor responses to antitumor drugs are variable, but predicting these responses is important when selecting effective chemotherapy treatments. Our aim was to identify variations or alterations in gene copy number that influence cancer cells’ susceptibilities to standard chemotherapeutic agents. Methods: Twenty-six human cancer cell lines representing the five main tumor types were subcutaneously implanted into nude mice and tested for sensitivity to fluorinated pyrimidines (UFT, TS-1, 5’-DFUR, and capecitabine), CDDP, CPT-11, and paclitaxel. The cell lines included lung (AOI, LC-11, Lu-99, LX-1, LC-6, Lu-134, Lu-130), colon (KM12C, KM12C/FU, HCT-15, COL-1, CO-3), pancreas (PAN-3, PAN-4, PAN-12, H-48, MIAPaCa-2, BxPC-3), gastric (SC-2, ST-40, 4-1ST, SC-4) and breast (MC-2, MX-1, MDA-MB-435SHM, MDA-MD-231). Genomic DNA was prepared from frozen tumor tissues. Mutations in 48 genes from the TruSeq Amplicon Cancer Panel were screened using the MiSeq system (Illumina, San Diego, CA). Somatic copy number alterations were analyzed by high-density SNP arrays (Affymetrix, Santa Clara, CA). Results: Of the 225 amplicons (187 non-overlapping regions) in the cancer panel, 86% achieved a minimum average sequencing depth of 1000X and the average coverage across all target regions was 5374X. In 26 tumors, sequencing detected 55 somatic mutations in 18 out of 48 cancer related genes of high prognostic or therapeutic significance, such as TP53, APC, PTEN, and SMAD4. Mutation frequencies across 26 xenografts were 73.1% (TP53), 38.5% (KRAS), 15.4% (APC), 11.5% (SMAD4 and RET), 7.7% (BRAF, GNAS, and PTEN), and 3.8% (CTNNB1, GNAQ, HNF1A, HRAS, IDH1, KIT, NOTCH1, PIK3CA, SMO, and STK11). Tumor xenografts with TP53 mutations were significantly less sensitive to CDDP and CPT-11 than wild-type cell lines (P Citation Format: Takashi Kobunai, Kenta Tsunekuni, Kazuaki Matsuoka, Hiroshi Tsukihara, Teiji Takechi. Mutational and copy number profiling of cancer-related genes in 26 human tumor xenografts and their correlations with antitumor drug sensitivities. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 5175.

Collaboration


Dive into the Kazuaki Matsuoka's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge