Network


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

Hotspot


Dive into the research topics where Tatsuya Fujikawa is active.

Publication


Featured researches published by Tatsuya Fujikawa.


BMC Cancer | 2009

Twist expression promotes migration and invasion in hepatocellular carcinoma.

Noriyuki Matsuo; Hidenori Shiraha; Tatsuya Fujikawa; Nobuyuki Takaoka; Naoki Ueda; Shigetomi Tanaka; Shinichi Nishina; Yutaka Nakanishi; Masayuki Uemura; Akinobu Takaki; Shinichiro Nakamura; Yoshiyuki Kobayashi; Kazuhiro Nouso; Takahito Yagi; Kazuhide Yamamoto

BackgroundTwist, a transcription factor of the basic helix-loop-helix class, is reported to regulate cancer metastasis. It is known to induce epithelial-mesenchymal transition (EMT). In this study, we evaluated the expression of twist and its effect on cell migration in hepatocellular carcinoma (HCC).MethodsWe examined twist expression using immunohistochemistry in 20 tissue samples of hepatocellular carcinoma, and assessed twist expression in HCC cell lines by RT-PCR and Western blot analysis. Ectopic twist expression was created by introducing a twist construct in the twist-negative HCC cell lines. Endogenous twist expression was blocked by twist siRNA in the twist-positive HCC cell lines. We studied EMT related markers, E-cadherin, Vimentin, and N-cadherin by Western blot analysis. Cell proliferation was measured by MTT assay, and cell migration was measured by in vitro wound healing assay. We used immunofluorescent vinculin staining to visualize focal adhesion.ResultsWe detected strong and intermediate twist expression in 7 of 20 tumor samples, and no significant twist expression was found in the tumor-free resection margins. In addition, we detected twist expression in HLE, HLF, and SK-Hep1 cells, but not in PLC/RPF/5, HepG2, and Huh7 cells. Ectopic twist-expressing cells demonstrated enhanced cell motility, but twist expression did not affect cell proliferation. Twist expression induced epithelial-mesenchymal transition together with related morphologic changes. Focal adhesion contact was reduced significantly in ectopic twist-expressing cells. Twist-siRNA-treated HLE, HLF, and SK-Hep1 cells demonstrated a reduction in cell migration by 50, 40 and 18%, respectively.ConclusionTwist induces migratory effect on hepatocellular carcinoma by causing epithelial-mesenchymal transition.


Journal of Biological Chemistry | 2005

Des-γ-carboxy Prothrombin Is a Potential Autologous Growth Factor for Hepatocellular Carcinoma

Mayumi Suzuki; Hidenori Shiraha; Tatsuya Fujikawa; Nobuyuki Takaoka; Naoki Ueda; Yutaka Nakanishi; Kazuko Koike; Akinobu Takaki; Yasushi Shiratori

Des-γ-carboxyl prothrombin (DCP) is a well recognized tumor marker for hepatocellular carcinoma (HCC). In the present study, we demonstrate that DCP has a mitogenic effect on HCC cell lines. Purified DCP stimulated DNA synthesis of Hep3B and SK-Hep-1 cells in a dose-dependent manner. DCP was found to bind with cell surface receptor Met causing Met autophosphorylation and also to activate STAT3 signaling pathway through Janus kinase 1. Luciferase gene reporter analysis showed that DCP induced STAT3-related transcription. Small interfering RNAs against both STAT3 and Met abrogated DCP-induced cell proliferation. DCP did not affect the mitogen-activated protein kinase pathway, Myc signaling pathway, or phosphoinositide 3-kinase/Akt pathway. Based on these results, we believe that DCP acts as an autologous mitogen for HCC cell lines. The Met-Janus kinase 1-STAT3 signaling pathway may be a major signaling pathway for DCP-induced cell proliferation.


Journal of Biological Chemistry | 2007

Des-γ-carboxyl Prothrombin-promoted Vascular Endothelial Cell Proliferation and Migration

Tatsuya Fujikawa; Hidenori Shiraha; Naoki Ueda; Nobuyuki Takaoka; Yutaka Nakanishi; Noriyuki Matsuo; Shigetomi Tanaka; Shin Ichi Nishina; Mayumi Suzuki; Akinobu Takaki; Kohsaku Sakaguchi; Yasushi Shiratori

Des-γ-carboxyl prothrombin (DCP) is a well recognized tumor marker for hepatocellular carcinoma. Previously, we have demonstrated that DCP stimulates cell proliferation in hepatocellular carcinoma cell lines through Met-Janus kinase 1 signal transducer and activator of transcription 3 signaling pathway. In the present study, we demonstrated that DCP induces both cell proliferation and migration in human umbilical vein endothelial cells. DCP was found to bind with the kinase insert domain receptor (KDR), alternatively referred to as vascular endothelial growth factor receptor-2. Furthermore, DCP induced autophosphorylation of KDR and its downstream effector phospholipase C-γ and mitogen-activated protein kinase (MAPK). To support these results, we showed that DCP-induced cell proliferation and cell migration were inhibited by KDR short interfering RNA, KDR kinase inhibitor, or MAPK inhibitor. In conclusion, these results indicate that DCP is a novel type of vascular endothelial growth factor that possesses potent mitogenic and migrative activities.


Journal of Gastroenterology and Hepatology | 2004

Telomerase reverse transcriptase gene amplification in hepatocellular carcinoma.

Yoshitaka Takuma; Kazuhiro Nouso; Yoshiyuki Kobayashi; Shinichiro Nakamura; Hironori Tanaka; Eiji Matsumoto; Tatsuya Fujikawa; Mayumi Suzuki; Tadashi Hanafusa; Yasushi Shiratori

Background and Aim:  Telomerase activation is essential for the immortality of cancer cells. The expression of telomerase reverse transcriptase (hTERT), the catalytic component of the telomerase complex, regulates telomerase activity in human cancers. Amplification of the hTERT gene, located at chromosome 5p, is thought to be a potential genetic event contributing to telomerase activation in sporadic tumors.


Journal of Biological Chemistry | 2012

Essential Roles of Raf/Extracellular Signal-regulated Kinase/Mitogen-activated Protein Kinase Pathway, YY1, and Ca2+ Influx in Growth Arrest of Human Vascular Smooth Muscle Cells by Bilirubin

Marlon Stoeckius; Anna Erat; Tatsuya Fujikawa; Makoto Hiromura; Anna Koulova; Leo E. Otterbein; Cesario Bianchi; Edda Tobiasch; Yossi Dagon; Frank W. Sellke; Anny Usheva

Background: Bilirubin circulates throughout the human cardiovascular system. Its interaction with the vascular wall is not well known. Results: Bilirubin alters Raf/ERK/MAPK pathway, cellular transcription factor YY1 location, and calcium-dependent YY1 proteolysis in human vascular cells. Conclusion: At high physiological levels bilirubin, inhibits cell growth, inhibits proliferation, and does not cause apoptosis. Significance: The observations provide opportunities for prevention and treatment of cardiovascular diseases. The biological effects of bilirubin, still poorly understood, are concentration-dependent ranging from cell protection to toxicity. Here we present data that at high nontoxic physiological concentrations, bilirubin inhibits growth of proliferating human coronary artery smooth muscle cells by three events. It impairs the activation of Raf/ERK/MAPK pathway and the cellular Raf and cyclin D1 content that results in retinoblastoma protein hypophosphorylation on amino acids S608 and S780. These events impede the release of YY1 to the nuclei and its availability to regulate the expression of genes and to support cellular proliferation. Moreover, altered calcium influx and calpain II protease activation leads to proteolytical degradation of transcription factor YY1. We conclude that in the serum-stimulated human vascular smooth muscle primary cell cultures, bilirubin favors growth arrest, and we propose that this activity is regulated by its interaction with the Raf/ERK/MAPK pathway, effect on cyclin D1 and Raf content, altered retinoblastoma protein profile of hypophosphorylation, calcium influx, and YY1 proteolysis. We propose that these activities together culminate in diminished 5 S and 45 S ribosomal RNA synthesis and cell growth arrest. The observations provide important mechanistic insight into the molecular mechanisms underlying the transition of human vascular smooth muscle cells from proliferative to contractile phenotype and the role of bilirubin in this transition.


Molecular Oncology | 2008

Exon 2 deletion splice variant of γ‐glutamyl carboxylase causes des‐γ‐carboxy prothrombin production in hepatocellular carcinoma cell lines

Naoki Ueda; Hidenori Shiraha; Tatsuya Fujikawa; Nobuyuki Takaoka; Yutaka Nakanishi; Mayumi Suzuki; Noriyuki Matsuo; Shigetomi Tanaka; Shin Ichi Nishina; Masayuki Uemura; Akinobu Takaki; Yasushi Shiratori; Kazuhide Yamamoto

Using GGCX gene‐specific real‐time PCR, exon 2 deletion splice variant of vitamin K‐dependent γ‐glutamyl carboxylase (GGCX) mRNA was identified in HCC cell lines. Expressions of wild type and exon 2 deletion variant of GGCX were analyzed with relevance to DCP production in HCC cell lines. Hep3B, HepG2, HuH1, HuH7, and PLC/PRF/5 produced DCP, while SK‐Hep‐1, HLE, HLF, and JHH1 produced no detectable level of DCP. DCP‐producing cells expressed exon 2 deletion variant of GGCX mRNA and protein, while DCP‐negative cells expressed no detectable level of exon 2 deletion variant of GGCX. These results suggest that exon 2 deletion splice variant of GGCX causes dysfunction of GGCX enzyme activity resulting in DCP production in HCC cell lines.


Liver International | 2004

Increased levels of tissue inhibitor of metalloproteinase-1 in human hepatocellular carcinoma.

Eiji Matsumoto; Harushige Nakatsukasa; Kazuhiro Nouso; Yoshiyuki Kobayashi; Shinichiro Nakamura; Mayumi Suzuki; Yoshitaka Takuma; Hironori Tanaka; Tatsuya Fujikawa; Yasushi Shiratori

Abstract: 
Background: Invasion and metastasis of hepatocellular carcinoma (HCC) are regulated by matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs). However, the role of TIMPs in these processes is not clear.


Journal of Gastroenterology and Hepatology | 2007

Cimetidine inhibits epidermal growth factor-induced cell signaling.

Tatsuya Fujikawa; Hidenori Shiraha; Yutaka Nakanishi; Nobuyuki Takaoka; Naoki Ueda; Mayumi Suzuki; Yasushi Shiratori

Background:  Cimetidine, a histamine‐2 (H2) receptor antagonist, has been demonstrated to have anticancer effects on colorectal cancer, melanoma and renal cell carcinoma. In the current study, we clarified that cimetidine inhibits both epidermal growth factor (EGF)‐induced cell proliferation and migration in hepatocellular carcinoma (HCC) cell lines.


Tumor Biology | 2006

High Level of Expression of α-Fetoprotein Receptor in Gastric Cancers

So Tsuboi; Kazuhisa Taketa; Kazuhiro Nouso; Tatsuya Fujikawa; Koji Manabe; Hiroyuki Ohmori; Toshihiro Higashi; Yasushi Shiratori

The expression of the receptor for α-fetoprotein (AFP-R) was examined immunohistochemically in 47 cancer and 14 benign human gastric tissues. Rabbit polyclonal antibody against human AFP-R was used for immunohistochemical staining. Thirty-four of the 47 cancer tissues expressed AFP-R showing granular or reticular staining on the cancer cell surface, while only 2 of 61 control cases (14 benign gastric tissues and 47 nonmalignant tissues adjacent to cancer) showed faint and homogeneous staining in the cytoplasm of noncancerous cells. There was a significant difference in staining intensity between the cancerous and noncancerous groups. However, no statistically significant difference in staining intensity was found among the groups of well-differentiated, moderately differentiated and poorly differentiated adenocarcinomas. On the other hand, the staining intensity of signet ring cell carcinoma was significantly weaker than that of the three adenocarcinoma groups. The high level of AFP-R expression in gastric cancers may allow the use of AFP-R as a new clinically useful marker of gastric cancer in the tissue level.


Comparative Hepatology | 2004

Elevated levels of tissue inhibitor of metalloproteinases (TIMPS) in human hepatocellular carcinomas

Eiji Matsumoto; Harushige Nakatsukasa; Kazuhiro Nouso; Shinichiro Nakamura; Mayumi Suzuki; Yoshiyuki Kobayashi; Masayuki Uemura; Syuichiro Sato; Eiichiro Yumoto; Junko Yokoyama; Sou Tsuboi; Hironori Tanaka; Yoshiataka Takuma; Tatsuya Fujikawa; Yasushi Shiratori

Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies in Japan. Invasion and metastasis of HCC are frequently observed during the development of HCCs, and are regulated by matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs). We have previously reported the strong expression of TIMP-1 and TIMP-2, and cellular localization of the transcripts in HCCs [1]. But the mechanisms of invasion and metastasis of HCCs, and particularly the role of TIMPs are still unclear. The purpose of this study was to examine the tissue concentration of the TIMP-1 and to study the potential involvement of TIMP-1 in the development of HCCs.

Collaboration


Dive into the Tatsuya Fujikawa'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