Network


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

Hotspot


Dive into the research topics where Kosuke Sakitani is active.

Publication


Featured researches published by Kosuke Sakitani.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Loss of liver E-cadherin induces sclerosing cholangitis and promotes carcinogenesis

Hayato Nakagawa; Yohko Hikiba; Yoshihiro Hirata; Joan Font-Burgada; Kei Sakamoto; Yoku Hayakawa; Koji Taniguchi; Atsushi Umemura; Hiroto Kinoshita; Kosuke Sakitani; Yuji Nishikawa; Kenji Hirano; Tsuneo Ikenoue; Hideaki Ijichi; Debanjan Dhar; Wataru Shibata; Masao Akanuma; Kazuhiko Koike; Michael Karin; Shin Maeda

Significance The precise roles of E-cadherin in the liver and liver carcinogenesis are still unknown. Here we show that mice lacking E-cadherin in the liver develop spontaneous periportal inflammation via an impaired intrahepatic biliary network, as well as periductal fibrosis, which resembles primary sclerosing cholangitis. Inducible gene knockout studies identified E-cadherin loss in biliary epithelial cells as a causal factor of cholangitis induction, and dysregulated E-cadherin expression was also seen in patients with primary sclerosing cholangitis. E-cadherin loss also significantly accelerates genetically and chemically engineered liver cancer through epithelial–mesenchymal transition, up-regulation of stem cell markers, and ERK activation. Thus, E-cadherin plays critical roles in maintaining homeostasis and suppressing carcinogenesis in the liver. E-cadherin is an important adhesion molecule whose loss is associated with progression and poor prognosis of liver cancer. However, it is unclear whether the loss of E-cadherin is a real culprit or a bystander in liver cancer progression. In addition, the precise role of E-cadherin in maintaining liver homeostasis is also still unknown, especially in vivo. Here we demonstrate that liver-specific E-cadherin knockout mice develop spontaneous periportal inflammation via an impaired intrahepatic biliary network, as well as periductal fibrosis, which resembles primary sclerosing cholangitis. Inducible gene knockout studies identified E-cadherin loss in biliary epithelial cells as a causal factor of cholangitis induction. Furthermore, a few of the E-cadherin knockout mice developed spontaneous liver cancer. When knockout of E-cadherin is combined with Ras activation or chemical carcinogen administration, E-cadherin knockout mice display markedly accelerated carcinogenesis and an invasive phenotype associated with epithelial–mesenchymal transition, up-regulation of stem cell markers, and elevated ERK activation. Also in human hepatocellular carcinoma, E-cadherin loss correlates with increased expression of mesenchymal and stem cell markers, and silencing of E-cadherin in hepatocellular carcinoma cell lines causes epithelial–mesenchymal transition and increased invasiveness, suggesting that E-cadherin loss can be a causal factor of these phenotypes. Thus, E-cadherin plays critical roles in maintaining homeostasis and suppressing carcinogenesis in the liver.


PLOS ONE | 2013

Interleukin-6 mediates epithelial-stromal interactions and promotes gastric tumorigenesis.

Hiroto Kinoshita; Yoshihiro Hirata; Hayato Nakagawa; Kei Sakamoto; Yoku Hayakawa; Ryota Takahashi; Wachiko Nakata; Kosuke Sakitani; Takako Serizawa; Yohko Hikiba; Masao Akanuma; Wataru Shibata; Shin Maeda; Kazuhiko Koike

Interleukin-6 (IL-6) is a pleiotropic cytokine that affects various functions, including tumor development. Although the importance of IL-6 in gastric cancer has been documented in experimental and clinical studies, the mechanism by which IL-6 promotes gastric cancer remains unclear. In this study, we investigated the role of IL-6 in the epithelial–stromal interaction in gastric tumorigenesis. Immunohistochemical analysis of human gastritis, gastric adenoma, and gastric cancer tissues revealed that IL-6 was frequently detected in the stroma. IL-6–positive cells in the stroma showed positive staining for the fibroblast marker α-smooth muscle actin, suggesting that stromal fibroblasts produce IL-6. We compared IL-6 knockout (IL-6−/−) mice with wild-type (WT) mice in a model of gastric tumorigenesis induced by the chemical carcinogen N-methyl-N-nitrosourea. The stromal fibroblasts expressed IL-6 in tumors from WT mice. Gastric tumorigenesis was attenuated in IL-6−/− mice, compared with WT mice. Impaired tumor development in IL-6−/− mice was correlated with the decreased activation of STAT3, a factor associated with gastric cancer cell proliferation. In vitro, when gastric cancer cell line was co-cultured with primary human gastric fibroblast, STAT3–related genes including COX-2 and iNOS were induced in gastric cancer cells and this response was attenuated with neutralizing anti-IL-6 receptor antibody. IL-6 production from fibroblasts was increased when fibroblasts were cultured in the presence of gastric cancer cell–conditioned media. IL-6 production from fibroblasts was suppressed by an interleukin-1 (IL-1) receptor antagonist and siRNA inhibition of IL-1α in the fibroblasts. IL-1α mRNA and protein were increased in fibroblast lysate, suggesting that cell-associated IL-1α in fibroblasts may be involved. Our results suggest the importance of IL-6 mediated stromal-epithelial cell interaction in gastric tumorigenesis.


Infection and Immunity | 2012

Role of Interleukin-32 in Helicobacter pylori-Induced Gastric Inflammation

Kosuke Sakitani; Yoshihiro Hirata; Yoku Hayakawa; Takako Serizawa; Wachiko Nakata; Ryota Takahashi; Hiroto Kinoshita; Kei Sakamoto; Hayato Nakagawa; Masao Akanuma; Haruhiko Yoshida; Shin Maeda; Kazuhiko Koike

ABSTRACT Helicobacter pylori infection is associated with gastritis and gastric cancer. An H. pylori virulence factor, the cag pathogenicity island (PAI), is related to host cell cytokine induction and gastric inflammation. Since elucidation of the mechanisms of inflammation is important for therapy, the associations between cytokines and inflammatory diseases have been investigated vigorously. Levels of interleukin-32 (IL-32), a recently described inflammatory cytokine, are increased in various inflammatory diseases, such as rheumatoid arthritis and Crohns disease, and in malignancies, including gastric cancer. In this report, we examined IL-32 expression in human gastric disease. We also investigated the function of IL-32 in activation of the inflammatory cytokines in gastritis. IL-32 expression paralleled human gastric tissue pathology, with low IL-32 expression in H. pylori-uninfected gastric mucosa and higher expression levels in gastritis and gastric cancer tissues. H. pylori infection increased IL-32 expression in human gastric epithelial cell lines. H. pylori-induced IL-32 expression was dependent on the bacterial cagPAI genes and on activation of nuclear factor κB (NF-κB). IL-32 expression induced by H. pylori was not detected in the supernatant of AGS cells but was found in the cytosol. Expression of the H. pylori-induced cytokines CXCL1, CXCL2, and IL-8 was decreased in IL-32-knockdown AGS cell lines compared to a control AGS cell line. We also found that NF-κB activation was decreased in H. pylori-infected IL-32-knockdown cells. These results suggest that IL-32 has important functions in the regulation of cytokine expression in H. pylori-infected gastric mucosa.


Cancer Science | 2012

Apoptosis signal-regulating kinase-1 inhibitor as a potent therapeutic drug for the treatment of gastric cancer.

Yoku Hayakawa; Yoshihiro Hirata; Kosuke Sakitani; Hayato Nakagawa; Wachiko Nakata; Hiroto Kinoshita; Ryota Takahashi; Kohsuke Takeda; Hidenori Ichijo; Shin Maeda; Kazuhiko Koike

Aside from the human epidermal growth factor receptor‐2 (HER2)‐targeting agent trastuzumab, molecular targeting therapy for gastric cancer (GC) has not been established. We previously reported that apoptosis signal‐regulating kinase‐1 (ASK1) was upregulated in human GC and that overexpression of ASK1 promoted GC cell proliferation. Here, we investigated the effect of ASK1 inhibitor K811 on GC cells. K811 efficiently prevented cell proliferation in cell lines with high ASK1 expression and in HER2‐overexpressing GC cells. Treatment with K811 reduced sizes of xenograft tumors by downregulating proliferation markers. These results indicate that ASK1 inhibition prevents GC cell growth in vitro and in vivo, suggesting that ASK1 inhibitors can be potent therapeutic drugs for GC.


Journal of Gastroenterology and Hepatology | 2011

Gastric cancer risk according to the distribution of intestinal metaplasia and neutrophil infiltration

Kosuke Sakitani; Yoshihiro Hirata; Hirotsugu Watabe; Atsuo Yamada; Takafumi Sugimoto; Yutaka Yamaji; Haruhiko Yoshida; Shin Maeda; Masao Omata; Kazuhiko Koike

Background and Aim:  Gastritis and intestinal metaplasia (IM) have long been known to be risk factors for and precursors of gastric cancer. We aimed to elucidate the association between gastric cancer risk and the distribution of precancerous lesions in the stomach by histological analyses.


Cancer Science | 2013

Therapeutic effect of c-Jun N-terminal kinase inhibition on pancreatic cancer

Ryota Takahashi; Yoshihiro Hirata; Kosuke Sakitani; Wachiko Nakata; Hiroto Kinoshita; Yoku Hayakawa; Hayato Nakagawa; Kei Sakamoto; Yohko Hikiba; Hideaki Ijichi; Harold L. Moses; Shin Maeda; Kazuhiko Koike

c‐Jun N‐terminal kinase (JNK) is a member of the mitogen‐activated protein kinase (MAPK) family, and it is reportedly involved in the development of several cancers. However, the role of JNK in pancreatic cancer has not been elucidated. We assessed t he involvement of JNK in the development of pancreatic cancer and investigated the therapeutic effect of JNK inhibitors on this deadly cancer. Small interfering RNAs against JNK or the JNK inhibitor SP600125 were used to examine the role of JNK in cellular proliferation and the cell cycles of pancreatic cancer cell lines. Ptf1acre/+;LSL‐KrasG12D/+;Tgfbr2flox/flox mice were treated with the JNK inhibitor to examine pancreatic histology and survival. The effect of JNK inhibition on tumor angiogenesis was also assessed using cell lines and murine pancreatic cancer specimens. JNK was frequently activated in human and murine pancreatic cancer in vitro and in vivo. Growth of human pancreatic cancer cell lines was suppressed by JNK inhibition through G1 arrest in the cell cycle with decreased cyclin D1 expression. In addition, oncogenic K‐ras expression led to activation of JNK in pancreatic cancer cell lines. Treatment of Ptf1acre/+;LSL‐KrasG12D/+;Tgfbr2flox/flox mice with the JNK inhibitor decreased growth of murine pancreatic cancer and prolonged survival of the mice significantly. Angiogenesis was also decreased by JNK inhibition in vitro and in vivo. In conclusion, activation of JNK promotes development of pancreatic cancer, and JNK may be a potential therapeutic target for pancreatic cancer.


Journal of Gastroenterology and Hepatology | 2015

Distribution of intestinal metaplasia as a predictor of gastric cancer development

Satoki Shichijo; Yoshihiro Hirata; Kosuke Sakitani; Shinzo Yamamoto; Takako Serizawa; Ryota Niikura; Hirotsugu Watabe; Shuntaro Yoshida; Atsuo Yamada; Yutaka Yamaji; Tetsuo Ushiku; Masashi Fukayama; Kazuhiko Koike

Helicobacter pylori, gastritis, and intestinal metaplasia (IM) are known risk factors for gastric cancer. In the present study, we conducted a cohort study to evaluate the predictive value of the distribution of IM for gastric cancer development.


International Journal of Antimicrobial Agents | 2012

Sitafloxacin resistance in Helicobacter pylori isolates and sitafloxacin-based triple therapy as a third-line regimen in Japan

Yoshihiro Hirata; Tomoya Ohmae; Ayako Yanai; Kosuke Sakitani; Yoku Hayakawa; Shuntaro Yoshida; Takafumi Sugimoto; Yuzo Mitsuno; Masao Akanuma; Yutaka Yamaji; Keiji Ogura; Shin Maeda; Kazuhiko Koike

The third-line treatment regimen for Helicobacter pylori after failure of clarithromycin- and metronidazole-based therapies is not yet established. Sitafloxacin (STX) is a quinolone that possesses potent in vitro activity against H. pylori. In this study, the susceptibility of H. pylori isolates to STX was examined and the efficacy of STX-based triple therapy as a third-line regimen was evaluated. STX showed minimum inhibitory concentrations (MICs) of ≤1 μg/mL against all 100 H. pylori isolates, and the MIC(90) (MIC for 90% of the organisms) of STX was 5 log(2) dilutions lower than that of levofloxacin (LVX). The MIC(50) (MIC for 50% of the organisms) of STX against gyrA mutants was 0.12 μg/mL and was significantly lower than that of LVX (8 μg/mL). The activity of STX at pH 5.5 was significantly less than that at pH 7.0. In the clinical trial, 28 patients with two eradication failures were treated with STX-based triple therapy [rabeprazole 10 mg twice daily (b.i.d.), amoxicillin 750 mg b.i.d. and STX 100mg b.i.d. for 7 days]. The eradication rate was 75% using intention-to-treat analysis and 80% using per-protocol analysis. Two gyrA mutant strains were eradicated. Amongst participants, a low pepsinogen I/II ratio was associated with successful eradication. These results suggest that STX could be active against most clinical H. pylori isolates and that STX-based triple therapy is a promising and safe third-line therapy.


Infection and Immunity | 2013

Differential roles of ASK1 and TAK1 in Helicobacter pylori-induced cellular responses

Yoku Hayakawa; Yoshihiro Hirata; Hiroto Kinoshita; Kosuke Sakitani; Hayato Nakagawa; Wachiko Nakata; Ryota Takahashi; Kei Sakamoto; Shin Maeda; Kazuhiko Koike

ABSTRACT The mitogen-activated protein kinase (MAPK) signaling pathway regulates various cellular functions, including those induced by Helicobacter pylori. TAK1 is an upstream MAPK kinase kinase (MAP3K) required for H. pylori-induced MAPK and NF-κB activation, but it remains unclear whether other MAP3Ks are involved in H. pylori-induced cellular responses. In this study, we focused on the MAP3K ASK1, which plays a critical role in gastric tumorigenesis. In gastric epithelial cells, H. pylori activates ASK1 in a reactive oxygen species (ROS)- and cag pathogenicity island-dependent manner, and ASK1 regulates sustained JNK activation and apoptosis induced by H. pylori. In contrast, TAK1 regulates H. pylori-mediated early JNK activation and cytokine production. We also found reciprocal regulation between ASK1 and TAK1 in H. pylori-related responses, whereby inhibition of TAK1 or downstream p38 MAPK activates ASK1 through ROS production, and ASK1 suppresses TAK1 and downstream NF-κB activation. We identified ROS/ASK1/JNK as a new signaling pathway induced by H. pylori, which regulates apoptotic cell death. The balance of ASK1-induced apoptosis and TAK1-induced antiapoptotic or inflammatory responses may determine the fate of epithelial cells infected with H. pylori and thus be involved in the pathogenesis of gastritis and gastric cancer.


Oncogene | 2013

Promotion of DNA repair by nuclear IKKβ phosphorylation of ATM in response to genotoxic stimuli.

Kei Sakamoto; Yohko Hikiba; Hayato Nakagawa; Yoshihiro Hirata; Yoku Hayakawa; Hiroto Kinoshita; Wachiko Nakata; Kosuke Sakitani; Ryota Takahashi; Masao Akanuma; Hideaki Kamata; Shin Maeda

Ataxia-telangiectasia mutated (ATM) is one of the key molecules involved in the cellular response to DNA damage. A portion of activated ATM is exported from the nucleus into the cytoplasm, where it activates the I kappa B kinase/nuclear factor kappa B (IKK/NF-κB) signaling pathway. It has been thought that activated IKKβ, which is a critical kinase for NF-κB activation, generally resides in the cytoplasm and phosphorylates cytoplasmic downstream molecules, such as IκBα. Here, we identified a new role for IKKβ during the response to DNA damage. ATM phosphorylation in response to alkylating agents consisted of two phases: the early phase (up to 3 h) and late phase (after 6 h). A portion of the activated IKKβ generated during the DNA damage response was found to translocate into the nucleus and directly phosphorylate ATM in the late phase. Furthermore, the phosphorylation of ATM by nuclear IKKβ was suggested to promote DNA repair. In parallel, activated IKKβ induced classical NF-κB activation and was involved in anti-apoptosis. Our findings define the function of IKKβ during the response to DNA damage, which promotes cell survival and DNA repair, and maintains cellular homeostasis.

Collaboration


Dive into the Kosuke Sakitani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shin Maeda

Yokohama City University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge