Network


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

Hotspot


Dive into the research topics where Sadahiko Kita is active.

Publication


Featured researches published by Sadahiko Kita.


Clinical Cancer Research | 2015

Keratin 19, a Cancer Stem Cell Marker in Human Hepatocellular Carcinoma

Takayuki Kawai; Kentaro Yasuchika; Takamichi Ishii; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Sadahiko Kita; Katsutaro Yasuda; Ken Fukumitsu; Masaki Mizumoto; Etsuro Hatano; Shinji Uemoto

Purpose: Keratin 19 (K19) is a known marker of poor prognosis and invasion in human hepatocellular carcinoma (HCC). However, the relationship between K19 and cancer stem cells (CSCs) is unclear. Here, we determined whether K19 can be used as a new CSC marker and therapeutic target in HCC. Experimental Design: HCC cell lines were transfected with a K19 promoter–driven enhanced green fluorescence protein gene. CSC characteristics, epithelial–mesenchymal transition (EMT), and TGFb/Smad signaling were examined in FACS-isolated K19+/K19− cells. K19 and TGFb receptor 1 (TGFbR1) expression in 166 consecutive human HCC surgical specimens was examined immunohistochemically. Results: FACS-isolated single K19+ cells showed self-renewal and differentiation into K19− cells, whereas single K19− cells did not produce K19+ cells. K19+ cells displayed high proliferation capacity and 5-fluorouracil resistance in vitro. Xenotransplantation into immunodeficient mice revealed that K19+ cells reproduced, differentiated into K19− cells, and generated large tumors at a high frequency in vivo. K19+ cells were found to be involved in EMT and the activation of TGFb/Smad signaling, and these properties were suppressed by K19 knockdown or treatment with a TGFbR1 inhibitor. The TGFbR1 inhibitor also showed high therapeutic effect against K19+ tumor in the mouse xenograft model. Immunohistochemistry of HCC specimens showed that compared with K19− patients, K19+ patients had significantly poorer recurrence-free survival and higher tumor TGFbR1 expression. Conclusions: K19 is a new CSC marker associated with EMT and TGFb/Smad signaling, and it would thus be a good therapeutic target for TGFbR1 inhibition. Clin Cancer Res; 21(13); 3081–91. ©2015 AACR.


Scientific Reports | 2016

SOX9 is a novel cancer stem cell marker surrogated by osteopontin in human hepatocellular carcinoma.

Takayuki Kawai; Kentaro Yasuchika; Takamichi Ishii; Yuya Miyauchi; Hidenobu Kojima; Ryoya Yamaoka; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Sadahiko Kita; Katsutaro Yasuda; Ken Fukumitsu; Junji Komori; Etsuro Hatano; Yoshiya Kawaguchi; Shinji Uemoto

The current lack of cancer stem cell (CSC) markers that are easily evaluated by blood samples prevents the establishment of new therapeutic strategies in hepatocellular carcinoma (HCC). Herein, we examined whether sex determining region Y-box 9 (SOX9) represents a new CSC marker, and whether osteopontin (OPN) can be used as a surrogate marker of SOX9 in HCC. In HCC cell lines transfected with a SOX9 promoter-driven enhanced green fluorescence protein gene, FACS-isolated SOX9+ cells were capable of self-renewal and differentiation into SOX9− cells, and displayed high proliferation capacity in vitro. Xenotransplantation experiments revealed that SOX9+ cells reproduced, differentiated into SOX9− cells, and generated tumors at a high frequency in vivo. Moreover, SOX9+ cells were found to be involved in epithelial-mesenchymal transition (EMT) and activation of TGFb/Smad signaling. Gain/loss of function experiments showed that SOX9 regulates Wnt/beta-catenin signaling, including cyclin D1 and OPN. Immunohistochemistry of 166 HCC surgical specimens and serum OPN measurements showed that compared to SOX9− patients, SOX9+ patients had significantly poorer recurrence-free survival, stronger venous invasion, and higher serum OPN levels. In conclusion, SOX9 is a novel HCC-CSC marker regulating the Wnt/beta-catenin pathway and its downstream target, OPN. OPN is a useful surrogate marker of SOX9 in HCC.


Scientific Reports | 2016

Efficient recellularisation of decellularised whole-liver grafts using biliary tree and foetal hepatocytes

Satoshi Ogiso; Kentaro Yasuchika; Ken Fukumitsu; Takamichi Ishii; Hidenobu Kojima; Yuya Miyauchi; Ryoya Yamaoka; Junji Komori; Hokahiro Katayama; Takayuki Kawai; E.Y. Yoshitoshi; Sadahiko Kita; Katsutaro Yasuda; Shinji Uemoto

A whole-organ regeneration approach, using a decellularised xenogeneic liver as a scaffold for the construction of a transplantable liver was recently reported. Deriving suitable scaffolds was the first step towards clinical application; however, effective recellularisation remains to be achieved. This report presents a strategy for the improvement of the recellularisation process, using novel cell-seeding technique and cell source. We evaluated recellularised liver grafts repopulated through the portal vein or the biliary duct with mice adult hepatocytes or E14.5 foetal hepatocytes. More than 80% of the cells seeded through the biliary tree entered the parenchyma beyond the ductule-lining matrix barrier and distributed throughout the liver lobule. In contrast, about 20% of the cells seeded through the portal tree entered the parenchyma. The gene expression levels of foetal hepatocyte albumin, glucose 6-phosphatase, transferrin, cytokeratin 19, and gamma-glutamyl transpeptidase were increased in three-dimensional cultures in the native liver-derived scaffolds, and the activation of liver detoxification enzymes and formation of biliary duct-like structures were supported. The metabolic functions of liver grafts recellularised with different cell types were similar. These results suggest that biliary tree cell-seeding approach is promising, and that liver progenitor cells represent a good cell source candidate.


Clinical Cancer Research | 2017

Identification of keratin19-positive cancer stem cells associating human hepatocellular carcinoma using 18F-fluorodeoxyglucose positron emission tomography.

Takayuki Kawai; Kentaro Yasuchika; Satoru Seo; Tatsuya Higashi; Takamichi Ishii; Yuya Miyauchi; Hidenobu Kojima; Ryoya Yamaoka; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Sadahiko Kita; Katsutaro Yasuda; Ken Fukumitsu; Yuji Nakamoto; Etsuro Hatano; Shinji Uemoto

Purpose: The current lack of tools for easy assessment of cancer stem cells (CSC) prevents the development of therapeutic strategies for hepatocellular carcinoma (HCC). We previously reported that keratin 19 (K19) is a novel HCC-CSC marker and that PET with 18F-fluorodeoxyglucose (18F-FDG) is an effective method for predicting postoperative outcome in hepatocellular carcinoma. Herein, we examined whether K19+ HCC-CSCs can be tracked using 18F-FDG-PET. Experimental Design: K19 and glucose transporter-1 (GLUT1) expression was evaluated by IHC in 98 hepatocellular carcinoma patients who underwent 18F-FDG-PET scans before primary tumor resection. Standardized uptake values (SUV) for primary tumors and tumor-to-nontumor SUV ratios (TNR) were calculated using FDG accumulation levels, and values were compared among K19+/K19− patients. Using hepatocellular carcinoma cell lines encoding with a K19 promoter–driven enhanced GFP, 18F-FDG uptake and GLUT1 expression were examined in FACS-isolated K19+/K19− cells. Results: In hepatocellular carcinoma patients, K19 expression was significantly correlated with GLUT1 expression and FDG accumulation. ROC analyses revealed that among preoperative clinical factors, TNR was the most sensitive indicator of K19 expression in hepatocellular carcinoma tumors. In hepatocellular carcinoma cells, FACS-isolated K19+ cells displayed significantly higher 18F-FDG uptake than K19− cells. Moreover, gain/loss-of-function experiments confirmed that K19 regulates 18F-FDG uptake through TGFβ/Smad signaling, including Sp1 and its downstream target GLUT1. Conclusions:18F-FDG-PET can be used to predict K19 expression in hepatocellular carcinoma and should thereby aid in the development of novel therapeutic strategies targeting K19+ HCC-CSCs. Clin Cancer Res; 23(6); 1450–60. ©2016 AACR.


American Journal of Transplantation | 2018

Establishment of practical recellularized liver graft for blood perfusion using primary rat hepatocytes and liver sinusoidal endothelial cells

Hidenobu Kojima; Kentaro Yasuchika; Ken Fukumitsu; Takamichi Ishii; Satoshi Ogiso; Yuya Miyauchi; Ryoya Yamaoka; Takayuki Kawai; Hokahiro Katayama; Elena Yukie Yoshitoshi-Uebayashi; Sadahiko Kita; Katsutaro Yasuda; Naoya Sasaki; Junji Komori; S. Uemoto

Tissue decellularization produces a three‐dimensional scaffold that can be used to fabricate functional liver grafts following recellularization. Inappropriate cell distribution and clotting during blood perfusion hinder the practical use of recellularized livers. Here we aimed to establish a seeding method for the optimal distribution of parenchymal and endothelial cells, and to evaluate the effect of liver sinusoidal endothelial cells (LSECs) in the decellularized liver. Primary rat hepatocytes and LSECs were seeded into decellularized whole‐liver scaffolds via the biliary duct and portal vein, respectively. Biliary duct seeding provided appropriate hepatocyte distribution into the parenchymal space, and portal vein–seeded LSECs simultaneously lined the portal lumen, thereby maintaining function and morphology. Hepatocytes co‐seeded with LSECs retained their function compared with those seeded alone. Platelet deposition was significantly decreased and hepatocyte viability was maintained in the co‐seeded group after extracorporeal blood perfusion. In conclusion, our seeding method provided optimal cell distribution into the parenchyma and vasculature according to the three‐dimensional structure of the decellularized liver. LSECs maintained hepatic function, and supported hepatocyte viability under blood perfusion in the engineered liver graft owing to their antithrombogenicity. This recellularization procedure could help produce practical liver grafts with blood perfusion.


Cell Transplantation | 2016

The Protective Effect of Transplanting Liver Cells Into the Mesentery on the Rescue of Acute Liver Failure After Massive Hepatectomy.

Sadahiko Kita; Kentaro Yasuchika; Takamichi Ishii; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Takayuki Kawai; Katsutaro Yasuda; Ken Fukumitsu; Masaki Mizumoto; Shinji Uemoto

Postoperative liver failure is one of the most critical complications following extensive hepatectomy. Although transplantation of allogeneic hepatocytes is an attractive therapy for posthepatectomy liver failure, transplanting cells via the portal veins typically causes portal vein embolization. The embolization by transplanted cells would be lethal in patients who have undergone massive hepatectomy. Thus, transplant surgeons need to select extrahepatic sites as transplant sites to prevent portal vein embolization. We aimed to investigate the mechanism of how liver cells transplanted into the mesentery protect recipient rats from acute liver failure after massive hepatectomy. We induced posthepatectomy liver failure by 90% hepatectomy in rats. Liver cells harvested from rat livers were transplanted into the mesenteries of hepatectomized rats. Twenty percent of the harvested cells, which consisted of hepatocytes and nonparenchymal cells, were transplanted into each recipient. The survival rate improved significantly in the liver cell transplantation group compared to the control group 7 days after hepatectomy (69 vs. 7%). Histological findings of the transplantation site, in vivo imaging system study findings, quantitative polymerase chain reaction assays of the transplanted cells, and serum albumin measurements of transplanted Nagase analbuminemic rats showed rapid deterioration of viable transplanted cells. Although viable transplanted cells deteriorated in the transplanted site, histological findings and an adenosine-5′-triphosphate (ATP) assay showed that the transplanted cells had a protective effect on the remaining livers. These results indicated that the paracrine effects of transplanted liver cells had therapeutic effects. The same protective effects were observed in the hepatocyte transplantation group, but not in the liver nonparenchymal cell transplantation group. Therefore, this effect on the remnant liver was mainly due to the hepatocytes among the transplanted liver cells. We demonstrated that transplanted liver cells protect the remnant liver from severe damage after massive hepatectomy.


Cancer Medicine | 2017

Identification of keratin 19-positive cancer stem cells associating human hepatocellular carcinoma using CYFRA 21-1

Takayuki Kawai; Kentaro Yasuchika; Takamichi Ishii; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Takahito Minami; Yuya Miyauchi; Hidenobu Kojima; Ryoya Yamaoka; Sadahiko Kita; Katsutaro Yasuda; Naoya Sasaki; Ken Fukumitsu; Etsuro Hatano; Shinji Uemoto

The current lack of an easily measurable surrogate marker of cancer stem cells (CSCs) prevents the clinical application of CSCs for hepatocellular carcinoma (HCC). We previously reported that keratin 19 (K19) is a novel HCC‐CSC marker associated with transforming growth factor beta (TGFβ)/Smad signaling, and that K19+ HCC‐CSCs could be a new therapeutic target of TGFβ receptor 1 inhibitor LY2157299. In this study, we examined whether K19+ HCC‐CSCs can be tracked using cytokeratin 19 fragment CYFRA 21‐1. In 147 HCC patients who underwent curative resection and evaluated K19 expression by immunohistochemistry, preoperative serum CYFRA 21‐1 levels were significantly higher in K19+ patients than in K19− patients (P < 0.01). Receiver operating characteristic analyses revealed that serum CYFRA 21‐1 was the statistically significant and the most sensitive predictor of tumor K19 expression among preoperative laboratory test values (P < 0.001). In HCC cells encoding with a K19 promoter‐driven enhanced green fluorescent protein, fluorescence‐activated cell sorting (FACS)‐isolated K19+ cells displayed significantly higher levels of supernatant CYFRA 21‐1 than K19− cells (P < 0.01). Gain/loss of K19 function experiments confirmed that CYFRA 21‐1 levels were regulated by K19 function in HCC cells. Furthermore, CYFRA 21‐1 levels reflected the treatment efficacy of LY2157299 in K19+ cells. In conclusion, CYFRA 21‐1 can be used to predict K19 expression in HCC, and should thereby aid in the development of novel therapeutic strategies targeting K19+ HCC‐CSCs.


Scientific Reports | 2017

Generation of non-viral, transgene-free hepatocyte like cells with piggyBac transposon

Hokahiro Katayama; Kentaro Yasuchika; Yuya Miyauchi; Hidenobu Kojima; Ryoya Yamaoka; Takayuki Kawai; E.Y. Yoshitoshi; Satoshi Ogiso; Sadahiko Kita; Katsutaro Yasuda; Naoya Sasaki; Ken Fukumitsu; Junji Komori; Takamichi Ishii; Shinji Uemoto

Somatic cells can be reprogrammed to induced hepatocyte-like cells (iHeps) by overexpressing certain defined factors in direct reprogramming techniques. Of the various methods to deliver genes into cells, typically used genome-integrating viral vectors are associated with integration-related adverse events such as mutagenesis, whereas non-integrating viral vectors have low efficiency, making viral vectors unsuitable for clinical application. Therefore, we focused on developing a transposon system to establish a non-viral reprogramming method. Transposons are unique DNA elements that can be integrated into and removed from chromosomes. PiggyBac, a type of transposon, has high transduction efficiency and cargo capacity, and the integrated transgene can be precisely excised in the presence of transposase. This feature enables the piggyBac vector to achieve efficient transgene expression and a transgene-free state, thus making it a promising method for cell reprogramming. Here, we attempted to utilize the piggyBac transposon system to generate iHeps by integrating a transgene consisting of Hnf4a and Foxa3, and successfully obtained functional iHeps. We then demonstrated removal of the transgene to obtain transgene-free iHeps, which still maintained hepatocyte functions. This non-viral, transgene-free reprogramming method using the piggyBac vector may facilitate clinical applications of iHeps in upcoming cell therapy.


Scientific Reports | 2017

A novel three-dimensional culture system maintaining the physiological extracellular matrix of fibrotic model livers accelerates progression of hepatocellular carcinoma cells

Yuya Miyauchi; Kentaro Yasuchika; Ken Fukumitsu; Takamichi Ishii; Satoshi Ogiso; Takahito Minami; Hidenobu Kojima; Ryoya Yamaoka; Hokahiro Katayama; Takayuki Kawai; Elena Yukie Yoshitoshi-Uebayashi; Sadahiko Kita; Katsutaro Yasuda; Naoya Sasaki; Shinji Uemoto

Liver fibrosis is characterized by the progressive accumulation of extracellular matrix (ECM) and is a strong predictor of hepatocellular carcinoma (HCC) development and progression. However, the effect of ECM in fibrotic livers on HCC cells is poorly understood. The aims of this study were to create a new culture system that retained the natural ECM of fibrotic model livers and to establish whether natural ECM regulated the characteristics of HCC cells. Using an organ decellularization technique, we created a new culture system that preserved the tissue-specific ECM of fibrotic model livers from CCl4-treated rats. The content of ECM in fibrotic model liver scaffolds was increased and the ECM microstructure was distorted. Quantitative polymerase chain reaction and immunofluorescence assays of HCC cells cultured in fibrotic model liver scaffolds for 7 days showed an epithelial-mesenchymal transition phenotype. Moreover, the ECM of fibrotic model livers promoted proliferation and chemoresistance of HCC cells. These results showed a novel effect of natural ECM in fibrotic model livers on the malignant behaviour of HCC cells. This new culture system will be useful for both understanding the cell biology of fibrotic livers and developing novel anti-cancer drugs.


Annals of Oncology | 2014

732PCYTOKERATIN 19, A CANCER STEM CELL MARKER IN HEPATOCELLULAR CARCINOMA

Takayuki Kawai; Kentaro Yasuchika; T. Ishii; Hokahiro Katayama; E.Y. Yoshitoshi; Satoshi Ogiso; Sadahiko Kita; Katsutaro Yasuda; Ken Fukumitsu; Masaki Mizumoto; Etsuro Hatano; Shinji Uemoto

ABSTRACT Aim: Cancer stem cells (CSCs) are receiving considerable attention as targets for cancer therapy. Although cytokeratin 19 (CK19) is known to involve in various malignant properties of hepatocellular carcinoma (HCC), the relationship between CK19 and CSCs is unclear. Here, we aimed to determine if CK19, a hepatic progenitor cell marker, can use as a new CSC marker in HCC. Methods: We examined CK19 expression of 166 consecutive human HCC specimens using immunohistochemistry, and analyzed the gene expression pattern of CK19+/CK19- patients. We transfected the CK19 promoter-driven enhanced green fluorescence protein gene into HCC cell lines, and investigated fluorescence-activated cell sorting (FACS)-isolated CK19+/CK19- cells. Results: CK19+ HCC patients had significantly poorer recurrence-free survival and larger tumor. They also showed the gene expression profile correlated with epithelial mesenchymal transition (EMT) and epithelial cell adhesion molecule (EpCAM). FACS-isolated single CK19+ cells showed the ability to self-renew and differentiate into CK19- cells, while single CK19- cells never produced CK19+ cells. CK19+ cells displayed greater proliferation capacity and stronger 5-fluorouracil resistance based on higher expression of multidrug-resistant protein 5 in vitro. Xenotransplantation into immunodeficiency mice revealed that CK19+ cells could reproduce themselves, differentiate into CK19- cells, and generate larger tumors at a higher frequency in vivo. CK19+ cells showed greater motility and higher expression of EMT-related genes than CK19- cells, and these properties were suppressed by transforming growth factor beta receptor 1 inhibitor LY2157299. CK19+ cells also expressed EpCAM at higher levels. Additionally, CK19+ cells had the advantage of greater proliferation potency over EpCAM. Conclusions: CK19+ cells are HCC-CSCs correlated with rapid tumor growth and EMT. CK19 should be a good therapeutic target. Disclosure: All authors have declared no conflicts of interest.

Collaboration


Dive into the Sadahiko Kita'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