Network


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

Hotspot


Dive into the research topics where Gouji Toyokawa is active.

Publication


Featured researches published by Gouji Toyokawa.


International Journal of Cancer | 2011

Dysregulation of PRMT1 and PRMT6, Type I arginine methyltransferases, is involved in various types of human cancers

Masanori Yoshimatsu; Gouji Toyokawa; Shinya Hayami; Motoko Unoki; Tatsuhiko Tsunoda; Helen I. Field; John D. Kelly; David E. Neal; Yoshihiko Maehara; Bruce A.J. Ponder; Yusuke Nakamura; Ryuji Hamamoto

Protein arginine methylation is a novel post‐translational modification regulating a diversity of cellular processes, including histone functions, but the roles of protein arginine methyltransferases (PRMTs) in human cancer are not well investigated. To address this issue, we first examined expression levels of genes belonging to the PRMT family and found significantly higher expression of PRMT1 and PRMT6, both of which are Type I PRMTs, in cancer cells of various tissues than in non‐neoplastic cells. Abrogation of the expression of these genes with specific siRNAs significantly suppressed growth of bladder and lung cancer cells. Expression profile analysis using the cells transfected with the siRNAs indicated that PRMT1 and PRMT6 interplay in multiple pathways, supporting regulatory roles in the cell cycle, RNA processing and also DNA replication that are fundamentally important for cancer cell proliferation. Furthermore, we demonstrated that serum asymmetric dimethylarginine (ADMA) levels of a number of cancer cases are significantly higher than those of nontumor control cases. In summary, our results suggest that dysregulation of PRMT1 and PRMT6 can be involved in human carcinogenesis and that these Type I arginine methyltransferases are good therapeutic targets for various types of cancer.


Cancer Research | 2011

Demethylation of RB regulator MYPT1 by histone demethylase LSD1 promotes cell cycle progression in cancer cells.

Hyun Soo Cho; Takehiro Suzuki; Naoshi Dohmae; Shinya Hayami; Motoko Unoki; Masanori Yoshimatsu; Gouji Toyokawa; Masashi Takawa; Taiping Chen; Julia K. Kurash; Helen I. Field; Bruce A.J. Ponder; Yusuke Nakamura; Ryuji Hamamoto

Histone demethylase LSD1 (also known as KDM1 and AOF2) is active in various cancer cells, but its biological significance in human carcinogenesis is unexplored. In this study, we explored hypothesized interactions between LSD1 and MYPT1, a known regulator of RB1 phosphorylation. We found that MYPT1 was methylated in vitro and in vivo by histone lysine methyltransferase SETD7 and demethylated by LSD1, identifying Lys 442 of MYPT1 as a target for methylation/demethylation by these enzymes. LSD1 silencing increased MYPT1 protein levels, decreasing the steady state level of phosphorylated RB1 (Ser 807/811) and reducing E2F activity. MYPT1 methylation status influenced the affinity of MYPT1 for the ubiquitin-proteasome pathway of protein turnover. MYPT1 was unstable in murine cells deficient in SETD7, supporting the concept that MYPT1 protein stability is physiologically regulated by methylation status. LSD1 overexpression could activate RB1 phosphorylation by inducing a destabilization of MYPT1 protein. Taken together, our results comprise a novel cell cycle regulatory mechanism mediated by methylation/demethylation dynamics, and they reveal the significance of LSD1 overexpression in human carcinogenesis.


Cancer Science | 2011

Validation of the histone methyltransferase EZH2 as a therapeutic target for various types of human cancer and as a prognostic marker

Masashi Takawa; Ken Masuda; Masaki Kunizaki; Yataro Daigo; Katsunori Takagi; Yukiko Iwai; Hyun-Soo Cho; Gouji Toyokawa; Yuka Yamane; Kazuhiro Maejima; Helen I. Field; Takaaki Kobayashi; Takayuki Akasu; Masanori Sugiyama; Eijyu Tsuchiya; Yutaka Atomi; Bruce A.J. Ponder; Yusuke Nakamura; Ryuji Hamamoto

The emphasis in anticancer drug discovery has always been on finding a drug with great antitumor potential but few side‐effects. This can be achieved if the drug is specific for a molecular site found only in tumor cells. Here, we find the enhancer of zeste homolog 2 (EZH2) to be highly overexpressed in lung and other cancers, and show that EZH2 is integral to proliferation in cancer cells. Quantitative real‐time PCR analysis revealed higher expression of EZH2 in clinical bladder cancer tissues than in corresponding non‐neoplastic tissues (P < 0.0001), and we confirmed that a wide range of cancers also overexpress EZH2, using cDNA microarray analysis. Immunohistochemical analysis showed positive staining for EZH2 in 14 of 29 cases of bladder cancer, 135 of 292 cases of non‐small‐cell lung cancer (NSCLC), and 214 of 245 cases of colorectal cancer, whereas no significant staining was observed in various normal tissues. We found elevated expression of EZH2 to be associated with poor prognosis for patients with NSCLC (P = 0.0239). In lung and bladder cancer cells overexpressing EZH2, suppression of EZH2 using specific siRNAs inhibited incorporation of BrdU and resulted in significant suppression of cell growth, even though no significant effect was observed in the normal cell strain CCD‐18Co, which has undetectable EZH2. Because EZH2 expression was scarcely detectable in all normal tissues we examined, EZH2 shows promise as a tumor‐specific therapeutic target. Furthermore, as elevated levels of EZH2 are associated with poor prognosis of patients with NSCLC, its overexpression in resected specimens could prove a useful molecular marker, indicating the necessity for a more extensive follow‐up in some lung cancer patients after surgical treatment. (Cancer Sci 2011; 102: 1298–1305)


Cancer Research | 2012

Histone Lysine Methyltransferase SETD8 Promotes Carcinogenesis by Deregulating PCNA Expression

Masashi Takawa; Hyun Soo Cho; Shinya Hayami; Gouji Toyokawa; Masaharu Kogure; Yuka Yamane; Yukiko Iwai; Kazuhiro Maejima; Koji Ueda; Akiko Masuda; Naoshi Dohmae; Helen I. Field; Tatsuhiko Tsunoda; Takaaki Kobayashi; Takayuki Akasu; Masanori Sugiyama; Shin Ichi Ohnuma; Yutaka Atomi; Bruce A.J. Ponder; Yusuke Nakamura; Ryuji Hamamoto

Although the physiologic significance of lysine methylation of histones is well known, whether lysine methylation plays a role in the regulation of nonhistone proteins has not yet been examined. The histone lysine methyltransferase SETD8 is overexpressed in various types of cancer and seems to play a crucial role in S-phase progression. Here, we show that SETD8 regulates the function of proliferating cell nuclear antigen (PCNA) protein through lysine methylation. We found that SETD8 methylated PCNA on lysine 248, and either depletion of SETD8 or substitution of lysine 248 destabilized PCNA expression. Mechanistically, lysine methylation significantly enhanced the interaction between PCNA and the flap endonuclease FEN1. Loss of PCNA methylation retarded the maturation of Okazaki fragments, slowed DNA replication, and induced DNA damage, and cells expressing a methylation-inactive PCNA mutant were more susceptible to DNA damage. An increase of methylated PCNA was found in cancer cells, and the expression levels of SETD8 and PCNA were correlated in cancer tissue samples. Together, our findings reveal a function for lysine methylation on a nonhistone protein and suggest that aberrant lysine methylation of PCNA may play a role in human carcinogenesis.


Nature Communications | 2012

Enhanced HSP70 lysine methylation promotes proliferation of cancer cells through activation of Aurora kinase B.

Hyun Soo Cho; Tadahiro Shimazu; Gouji Toyokawa; Yataro Daigo; Yoshihiko Maehara; Shinya Hayami; Akihiro Ito; Ken Masuda; Noriko Ikawa; Helen I. Field; Eiju Tsuchiya; Shin Ichi Ohnuma; Bruce A.J. Ponder; Minoru Yoshida; Yusuke Nakamura; Ryuji Hamamoto

Although heat-shock protein 70 (HSP70), an evolutionarily highly conserved molecular chaperone, is known to be post-translationally modified in various ways such as phosphorylation, ubiquitination and glycosylation, physiological significance of lysine methylation has never been elucidated. Here we identify dimethylation of HSP70 at Lys-561 by SETD1A. Enhanced HSP70 methylation was detected in various types of human cancer by immunohistochemical analysis, although the methylation was barely detectable in corresponding non-neoplastic tissues. Interestingly, methylated HSP70 predominantly localizes to the nucleus of cancer cells, whereas most of the HSP70 protein locates to the cytoplasm. Nuclear HSP70 directly interacts with Aurora kinase B (AURKB) in a methylation-dependent manner and promotes AURKB activity in vitro and in vivo. We also find that methylated HSP70 has a growth-promoting effect in cancer cells. Our findings demonstrate a crucial role of HSP70 methylation in human carcinogenesis.


Molecular Cancer | 2011

Minichromosome Maintenance Protein 7 is a potential therapeutic target in human cancer and a novel prognostic marker of non-small cell lung cancer

Gouji Toyokawa; Ken Masuda; Yataro Daigo; Hyun-Soo Cho; Masanori Yoshimatsu; Masashi Takawa; Shinya Hayami; Kazuhiro Maejima; Makoto Chino; Helen I. Field; David E. Neal; Eiju Tsuchiya; Bruce A.J. Ponder; Yoshihiko Maehara; Yusuke Nakamura; Ryuji Hamamoto

BackgroundThe research emphasis in anti-cancer drug discovery has always been to search for a drug with the greatest antitumor potential but fewest side effects. This can only be achieved if the drug used is against a specific target located in the tumor cells. In this study, we evaluated Minichromosome Maintenance Protein 7 (MCM7) as a novel therapeutic target in cancer.ResultsImmunohistochemical analysis showed that MCM7 was positively stained in 196 of 331 non-small cell lung cancer (NSCLC), 21 of 29 bladder tumor and 25 of 70 liver tumor cases whereas no significant staining was observed in various normal tissues. We also found an elevated expression of MCM7 to be associated with poor prognosis for patients with NSCLC (P = 0.0055). qRT-PCR revealed a higher expression of MCM7 in clinical bladder cancer tissues than in corresponding non-neoplastic tissues (P < 0.0001), and we confirmed that a wide range of cancers also overexpressed MCM7 by cDNA microarray analysis. Suppression of MCM7 using specific siRNAs inhibited incorporation of BrdU in lung and bladder cancer cells overexpressing MCM7, and suppressed the growth of those cells more efficiently than that of normal cell strains expressing lower levels of MCM7.ConclusionsSince MCM7 expression was generally low in a number of normal tissues we examined, MCM7 has the characteristics of an ideal candidate for molecular targeted cancer therapy in various tumors and also as a good prognostic biomarker for NSCLC patients.


International Journal of Cancer | 2012

The JmjC domain-containing histone demethylase KDM3A is a positive regulator of the G1/S transition in cancer cells via transcriptional regulation of the HOXA1 gene.

Hyun Soo Cho; Gouji Toyokawa; Yataro Daigo; Shinya Hayami; Ken Masuda; Noriko Ikawa; Yuka Yamane; Kazuhiro Maejima; Tatsuhiko Tsunoda; Helen I. Field; John D. Kelly; David E. Neal; Bruce A.J. Ponder; Yoshihiko Maehara; Yusuke Nakamura; Ryuji Hamamoto

A number of histone demethylases have been identified and biochemically characterized, yet their biological functions largely remain uncharacterized, particularly in the context of human diseases such as cancer. In this study, we describe important roles for the histone demethylase KDM3A, also known as JMJD1A, in human carcinogensis. Expression levels of KDM3A were significantly elevated in human bladder carcinomas compared with nonneoplastic bladder tissues (p < 0.0001), when assessed by real‐time PCR. We confirmed that some other cancers including lung cancer also overexpressed KDM3A, using cDNA microarray analysis. Treatment of cancer cell lines with small interfering RNA targeting KDM3A significantly knocked down its expression and resulted in the suppression of proliferation. Importantly, we found that KDM3A activates transcription of the HOXA1 gene through demethylating histone H3 at lysine 9 di‐methylation by binding to its promoter region. Indeed, expression levels of KDM3A and HOXA1 in several types of cancer cell lines and bladder cancer samples were statistically correlated. We observed the down‐regulation of HOXA1 as well as CCND1 after treatment with KDM3A siRNA, indicating G1 arrest of cancer cells. Together, our results suggest that elevated expression of KDM3A plays a critical role in the growth of cancer cells, and further studies may reveal a cancer therapeutic potential in KDM3A inhibition.


Journal of Thoracic Oncology | 2016

Clinical Significance of PD-L1 Protein Expression in Surgically Resected Primary Lung Adenocarcinoma.

Kazuki Takada; Tatsuro Okamoto; Fumihiro Shoji; Mototsugu Shimokawa; Takaki Akamine; Shinkichi Takamori; Masakazu Katsura; Yuzo Suzuki; Takatoshi Fujishita; Gouji Toyokawa; Yosuke Morodomi; Shinji Okano; Yoshinao Oda; Yoshihiko Maehara

Introduction The clinicopathological features of carcinomas expressing programmed death ligand 1 (PD‐L1) and their associations with common driver mutations, such as mutations in the EGFR gene, in lung adenocarcinoma are not clearly understood. Here, we examined PD‐L1 protein expression in surgically resected primary lung adenocarcinoma and the association of PD‐L1 protein expression with clinicopathological features, EGFR mutation status, and patient outcomes. Methods The expression of PD‐L1 protein in 417 surgically resected primary lung adenocarcinomas was evaluated by immunohistochemical analysis. The cutoff value for defining PD‐L1 positivity was determined according to the histogram of proportions of PD‐L1–positive cancer cells. Results Samples from 85 patients (20.4%) and 144 patients (34.5%) were positive for PD‐L1 protein expression according to 5% and 1% PD‐L1 cutoff values, respectively. Fisher’s exact tests showed that PD‐L1 positivity was significantly associated with male sex, smoking, higher tumor grade, advanced T status, advanced N status, advanced stage, the presence of pleural and vessel invasions, micropapillary or solid predominant histological subtypes, and wild‐type EGFR. Univariate and multivariate survival analyses revealed that patients with PD‐L1 positivity had poorer prognoses than those without PD‐L1 protein expression at the 1% cutoff value (disease‐free survival p < 0.0001, overall survival p < 0.0001). Conclusions PD‐L1 protein expression was significantly higher in smoking‐associated adenocarcinoma and in EGFR mutation–negative adenocarcinoma. PD‐L1 protein expression was associated with poor survival in patients with lung adenocarcinoma. The PD‐L1/programmed cell death 1 pathway may contribute to the progression of smoking‐associated tumors in lung adenocarcinoma.


Cancer Prevention Research | 2011

The Histone Demethylase JMJD2B Plays an Essential Role in Human Carcinogenesis through Positive Regulation of Cyclin-Dependent Kinase 6

Gouji Toyokawa; Hyun-Soo Cho; Yukiko Iwai; Masanori Yoshimatsu; Masashi Takawa; Shinya Hayami; Kazuhiro Maejima; Noriaki Shimizu; Hirotoshi Tanaka; Tatsuhiko Tsunoda; Helen I. Field; John Kelly; David E. Neal; Bruce A.J. Ponder; Yoshihiko Maehara; Yusuke Nakamura; Ryuji Hamamoto

Histone methyltransferases and demethylases are known to regulate transcription by altering the epigenetic marks on histones, but the pathologic roles of their dysfunction in human diseases, such as cancer, still remain to be elucidated. Herein, we show that the histone demethylase JMJD2B is involved in human carcinogenesis. Quantitative real-time PCR showed notably elevated levels of JMJD2B expression in bladder cancers, compared with corresponding nonneoplastic tissues (P < 0.0001), and elevated protein expression was confirmed by immunohistochemistry. In addition, cDNA microarray analysis revealed transactivation of JMJD2B in lung cancer, and immunohistochemical analysis showed protein overexpression in lung cancer. siRNA-mediated reduction of expression of JMJD2B in bladder and lung cancer cell lines significantly suppressed the proliferation of cancer cells, and suppressing JMJD2B expression lead to a decreased population of cancer cells in S phase, with a concomitant increase of cells in G1 phase. Furthermore, a clonogenicity assay showed that the demethylase activity of JMJD2B possesses an oncogenic activity. Microarray analysis after knockdown of JMJD2B revealed that JMJD2B could regulate multiple pathways which contribute to carcinogenesis, including the cell-cycle pathway. Of the downstream genes, chromatin immunoprecipitation showed that CDK6 (cyclin-dependent kinase 6), essential in G1–S transition, was directly regulated by JMJD2B, via demethylation of histone H3-K9 in its promoter region. Expression levels of JMJD2B and CDK6 were significantly correlated in various types of cell lines. Deregulation of histone demethylation resulting in perturbation of the cell cycle, represents a novel mechanism for human carcinogenesis and JMJD2B is a feasible molecular target for anticancer therapy. Cancer Prev Res; 4(12); 2051–61. ©2011 AACR.


Cancer Letters | 2013

Deregulation of the histone demethylase JMJD2A is involved in human carcinogenesis through regulation of the G1/S transition

Masaharu Kogure; Masashi Takawa; Hyun Soo Cho; Gouji Toyokawa; Kazuyuki Hayashi; Tatsuhiko Tsunoda; Takaaki Kobayashi; Yataro Daigo; Masanori Sugiyama; Yutaka Atomi; Yusuke Nakamura; Ryuji Hamamoto

Although a number of JmjC-containing histone demethylases have been identified and biochemically characterized, pathological roles of their dysfunction in human disease such as cancer have not been well elucidated. Here, we report the Jumonji domain containing 2A (JMJD2A) is integral to proliferation of cancer cells. Quantitative real-time PCR analysis revealed higher expression of JMJD2A in clinical bladder cancer tissues than in corresponding non-neoplastic tissues (P<0.0001). Immunohistochemical analysis also showed positive staining for JMJD2A in 288 out of 403 lung cancer cases, whereas no staining was observed in lung normal tissues. Suppression of JMJD2A expression in lung and bladder cancer cells overexpressing this gene, using specific siRNAs, inhibited incorporation of BrdU and resulted in significant suppression of cell growth. Furthermore, JMJD2A appears to directly transactivate the expression of some tumor associated proteins including ADAM12 through the regulation of histone H3K9 methylation. As expression levels of JMJD2A are low in normal tissues, it may be feasible to develop specific inhibitors targeting the enzyme as anti-tumor agents which should have a minimal risk of adverse reaction.

Collaboration


Dive into the Gouji Toyokawa'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