Network


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

Hotspot


Dive into the research topics where Masayuki Koizumi is active.

Publication


Featured researches published by Masayuki Koizumi.


Nature Genetics | 2011

Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine

Kenichiro Furuyama; Yoshiya Kawaguchi; Haruhiko Akiyama; Masashi Horiguchi; S. Kodama; T. Kuhara; Shinichi Hosokawa; Ashraf Elbahrawy; Tsunemitsu Soeda; Masayuki Koizumi; Toshihiko Masui; Michiya Kawaguchi; Kyoichi Takaori; Ryuichiro Doi; Eiichiro Nishi; Ryosuke Kakinoki; Jian Min Deng; Richard R. Behringer; Takashi Nakamura; Shinji Uemoto

The liver and exocrine pancreas share a common structure, with functioning units (hepatic plates and pancreatic acini) connected to the ductal tree. Here we show that Sox9 is expressed throughout the biliary and pancreatic ductal epithelia, which are connected to the intestinal stem-cell zone. Cre-based lineage tracing showed that adult intestinal cells, hepatocytes and pancreatic acinar cells are supplied physiologically from Sox9-expressing progenitors. Combination of lineage analysis and hepatic injury experiments showed involvement of Sox9-positive precursors in liver regeneration. Embryonic pancreatic Sox9-expressing cells differentiate into all types of mature cells, but their capacity for endocrine differentiation diminishes shortly after birth, when endocrine cells detach from the epithelial lining of the ducts and form the islets of Langerhans. We observed a developmental switch in the hepatic progenitor cell type from Sox9-negative to Sox9-positive progenitors as the biliary tree develops. These results suggest interdependence between the structure and homeostasis of endodermal organs, with Sox9 expression being linked to progenitor status.


Clinical Cancer Research | 2004

N-Cadherin Expression and Epithelial-Mesenchymal Transition in Pancreatic Carcinoma

Sanae Nakajima; Ryuichiro Doi; Eiji Toyoda; Shoichiro Tsuji; Michihiko Wada; Masayuki Koizumi; Sidhartha Tulachan; Daisuke Ito; Kazuhiro Kami; Tomohiko Mori; Yoshiya Kawaguchi; Koji Fujimoto; Ryo Hosotani; Masayuki Imamura

Purpose: Loss of intercellular adhesion and increased cell motility promote tumor cell invasion. In the present study, E- and N-cadherin, members of the classical cadherin family, are investigated as inducers of epithelial-to-mesenchymal transition (EMT) that is thought to play a fundamental role during the early steps of invasion and metastasis of carcinomas. Cell growth factors are known to regulate cell adhesion molecules. The purpose of the study presented here was to investigate whether a gain in N-cadherin in pancreatic cancer is involved in the process of metastasis via EMT and whether its expression is affected by growth factors. Experimental Design: We immunohistochemically examined the expression of N- and E-cadherins and vimentin, a mesenchymal marker, in pancreatic primary and metastatic tumors. Correlations among the expressions of N-cadherin, transforming growth factor (TGF)β, and fibroblast growth factor 2 was evaluated in both tumors, and the induction of cadherin and vimentin by growth factors was examined in cultured cell lines. Results: N-cadherin expression was observed in 13 of 30 primary tumors and in 8 of 15 metastatic tumors. N-cadherin expression correlated with neural invasion (P = 0.008), histological type (P = 0.043), fibroblast growth factor expression in primary tumors (P = 0.007), and TGF expression (P = 0.004) and vimentin (P = 0.01) in metastatic tumors. Vimentin, a mesenchymal marker, was observed in a few cancer cells of primary tumor but was substantially expressed in liver metastasis. TGF stimulated N-cadherin and vimentin protein expression and decreased E-cadherin expression of Panc-1 cells with morphological change. Conclusion: This study provided the morphological evidence of EMT in pancreatic carcinoma and revealed that overexpression of N-cadherin is involved in EMT and is affected by growth factors.


Journal of Clinical Investigation | 2006

Ectopic pancreas formation in Hes1 -knockout mice reveals plasticity of endodermal progenitors of the gut, bile duct, and pancreas

Akihisa Fukuda; Yoshiya Kawaguchi; Kenichiro Furuyama; S. Kodama; Masashi Horiguchi; T. Kuhara; Masayuki Koizumi; Daniel F. Boyer; Koji Fujimoto; Ryuichiro Doi; Ryoichiro Kageyama; Christopher V.E. Wright; Tsutomu Chiba

Ectopic pancreas is a developmental anomaly occasionally found in humans. Hes1, a main effector of Notch signaling, regulates the fate and differentiation of many cell types during development. To gain insights into the role of the Notch pathway in pancreatic fate determination, we combined the use of Hes1-knockout mice and lineage tracing employing the Cre/loxP system to specifically mark pancreatic precursor cells and their progeny in Ptf1a-cre and Rosa26 reporter mice. We show that inactivation of Hes1 induces misexpression of Ptf1a in discrete regions of the primitive stomach and duodenum and throughout the common bile duct. All ectopic Ptf1a-expressing cells were reprogrammed, or transcommitted, to multipotent pancreatic progenitor status and subsequently differentiated into mature pancreatic exocrine, endocrine, and duct cells. This process recapitulated normal pancreatogenesis in terms of morphological and genetic features. Furthermore, analysis of Hes1/Ptf1a double mutants revealed that ectopic Ptf1a-cre lineage-labeled cells adopted the fate of region-appropriate gut epithelium or endocrine cells similarly to Ptf1a-inactivated cells in the native pancreatic buds. Our data demonstrate that the Hes1-mediated Notch pathway is required for region-appropriate specification of pancreas in the developing foregut endoderm through regulation of Ptf1a expression, providing novel insight into the pathogenesis of ectopic pancreas development in a mouse model.


International Journal of Cancer | 2006

In vivo antitumor effect of the mTOR inhibitor CCI‐779 and gemcitabine in xenograft models of human pancreatic cancer

Daisuke Ito; Koji Fujimoto; Tomohiko Mori; Kazuhiro Kami; Masayuki Koizumi; Eiji Toyoda; Yoshiya Kawaguchi; Ryuichiro Doi

Mammalian target of rapamycin (mTOR) is considered to be a major effector of cell growth and proliferation that controls protein synthesis through a large number of downstream targets. We investigated the expression of the phosphatidylinositol 3′‐kinase (PI3K)/mTOR signaling pathway in human pancreatic cancer cells and tissues, and the in vivo antitumor effects of the mTOR inhibitor CCI‐779 with/without gemcitabine in xenograft models of human pancreatic cancer. We found that the Akt, mTOR and p70 S6 kinase (S6K1) from the PI3K/mTOR signaling pathway were activated in all of the pancreatic cancer cell lines examined. When surgically resected tissue specimens of pancreatic ductal adenocarcinoma were examined, phosphorylation of Akt, mTOR and S6K1 was detected in 50, 55 and 65% of the specimens, respectively. Although CCI‐779 had no additive or synergistic antiproliferative effect when combined with gemcitabine in vitro, it showed significant antitumor activity in the AsPC‐1 subcutaneous xenograft model as both a single agent and in combination with gemictabine. Furthermore, in the Suit‐2 peritoneal dissemination xenograft model, the combination of these 2 drugs achieved significantly better survival when compared with CCI‐779 or gemcitabine alone. These results demonstrate promising activity of the mTOR inhibitor CCI‐779 against human pancreatic cancer, and suggest that the inhibition of mTOR signaling can be exploited as a potentially tumor‐selective therapeutic strategy.


Journal of Hepato-biliary-pancreatic Surgery | 2009

Single-institution validation of the international consensus guidelines for treatment of branch duct intraductal papillary mucinous neoplasms of the pancreas

Kazuyuki Nagai; Ryuichiro Doi; Tatsuo Ito; Atsushi Kida; Masayuki Koizumi; Toshihiko Masui; Yoshiya Kawaguchi; Kohei Ogawa; Shinji Uemoto

BACKGROUND The international consensus guidelines (the guidelines) for management of intraductal papillary mucinous neoplasms (IPMNs) of the pancreas recommend surgical resection of branch duct IPMNs with any of the following features: cyst size >30 mm, mural nodules, main pancreatic duct diameter >6 mm, positive cytology, and symptoms. The aim of this study was to evaluate the usefulness of these guidelines for resection of branch duct IPMNs. METHODS We reviewed 84 consecutive patients with branch duct IPMNs who underwent surgical resection at our hospital between January 1984 and December 2007. RESULTS Sixty-nine patients had indications for resection according to the guidelines. Malignant IPMNs had significantly larger cysts than benign tumors (P = 0.026). Patients with malignant IPMNs had significantly more indications for resection than those with benign IPMNs (2.6 +/- 1.0 vs. 1.7 +/- 0.9, P < 0.001), and 36 of the 37 patients with malignant IPMNs had indications. The sensitivity of the guidelines for predicting malignancy was 97.3%. One of 15 patients without indications had malignancy, and the specificity was low (29.8%). CONCLUSIONS The guidelines show a high sensitivity for predicting malignancy of branch duct IPMNs, but the specificity is low. The cyst size and the total number of indications in each patient should be taken into account when predicting the risk of malignancy for branch duct IPMNs.


International Journal of Cancer | 2003

Endogenous decoy receptor 3 blocks the growth inhibition signals mediated by Fas ligand in human pancreatic adenocarcinoma

Shoichiro Tsuji; Ryo Hosotani; Shin Yonehara; Toshihiko Masui; Sidhartha Tulachan; Sanae Nakajima; Hiroyuki Kobayashi; Masayuki Koizumi; Eiji Toyoda; Daisuke Ito; Kazuhiro Kami; Tomohiko Mori; Koji Fujimoto; Ryuichiro Doi; Masayuki Imamura

Many cancers are resistant to Fas‐mediated apoptosis despite the expression of Fas. To investigate the mechanisms by which Fas signals are attenuated, we focused on decoy receptor 3 (DcR3). DcR3 is a soluble receptor against Fas ligand belonging to the tumor necrosis factor receptor superfamily and overexpresses in some forms of cancers. Exogenous DcR3 inhibits Fas‐mediated apoptosis in Fas‐sensitive Jurkat cells. In our study, we examined the expression and function of DcR3 in pancreatic cancers. TaqMan RT‐PCR showed that DcR3 mRNA was highly expressed in pancreatic cancer cell lines (71%) and tissues (67%). Its expression significantly correlated with cancer invasion to veins. Western blotting showed that the DcR3 protein was produced and secreted in 4 of 6 cell lines. The protein expressions were compatible with the mRNA expression. Five of 7 pancreatic cancer cell lines became sensitive to agonistic anti‐Fas antibody (CH‐11) to various extents, without Fas upregulation, when exposed to CH‐11 for 48 hr after pretreatment with IFNγ. Four of 7 pancreatic cancer cell lines were inhibited from growing, compared to control cells, when cocultured with membrane‐bounded Fas ligand (mFasL) transfected lymphomas for 48 hr after pretreatment with IFNγ. DcR3 reduced this growth inhibition when added exogenously. Regression analysis showed that the DcR3 expression significantly correlated with the sensitivity to mFasL, and not to CH‐11. These results suggest that DcR3 is highly expressed in many pancreatic cancers and endogenous DcR3 blocks the growth inhibition signals mediated by mFasL. DcR3 can be a candidate target molecule for the therapeutic intervention.


World Journal of Surgery | 2006

Clinical Significance of Focal Adhesion Kinase in Resectable Pancreatic Cancer

Kenichiro Furuyama; Ryuichiro Doi; Tomohiko Mori; Eiji Toyoda; Daisuke Ito; Kazuhiro Kami; Masayuki Koizumi; Atsushi Kida; Yoshiya Kawaguchi; Koji Fujimoto

Focal adhesion kinase (FAK) is a non-receptor, cytoplasmic protein tyrosine kinase that is involved in the regulation of cellular signaling, migration, apoptosis, and cell cycle progression. Previous reports have shown that FAK is expressed in various kinds of cancer tissues and cancer cell lines; however, no information is available about human pancreatic carcinoma specimens. Tissue such specimens were obtained from 50 patients who underwent pancreatic resection for pancreatic invasive ductal carcinoma at our institute from 1996 to 2002. Immunohistochemical analysis of FAK was performed in the resected specimens. Focal adhesion kinase expression in seven human pancreatic cancer cell lines was analyzed by reverse transcription polymerase chain reaction (PCR) analysis and Western blot analysis. Focal adhesion kinase expression was detected in 24 of 50 cases (48%). There was a statistically significant correlation between FAK expression and tumor size (P = 0.004), although FAK expression did not significantly correlate with other factors such as tumor histological grade, lymph node metastasis, distant metastasis, histological stage, and overall survival. Reverse transcription PCR analysis and Western blot analysis showed that FAK was expressed in all seven pancreatic cancer cell lines. Focal adhesion kinase expression was not directly related to clinicopathological factors except tumor size in pancreatic carcinoma. Focal adhesion kinase expression may not be a prognostic marker for pancreatic cancer patients.


Journal of Experimental & Clinical Cancer Research | 2009

Prognostic value of metastin expression in human pancreatic cancer

Kazuyuki Nagai; Ryuichiro Doi; Fumihiko Katagiri; Tatsuo Ito; Atsushi Kida; Masayuki Koizumi; Toshihiko Masui; Yoshiya Kawaguchi; Kenji Tomita; Shinya Oishi; Nobutaka Fujii; Shinji Uemoto

BackgroundKiSS-1 was identified as a metastasis-suppressing gene in melanoma cells. The KiSS-1 gene product (metastin) was isolated from human placenta as the ligand of GPR54, a G-protein-coupled receptor. The role of metastin and GPR54 in tumor progression is not fully understood.MethodsWe investigated the clinical significance of metastin and GPR54 expression in pancreatic cancer. We evaluated immunohistochemical expression of metastin and GPR54 in pancreatic ductal adenocarcinoma tissues obtained from 53 consecutive patients who underwent resection between July 2003 and May 2007 at Kyoto University Hospital. In 23 consecutive patients, the plasma metastin level was measured before surgery by enzyme immunoassay.ResultsStrong immunohistochemical expression of metastin was detected in 13 tumors (24.5%), while strong expression of GPR54 was detected in 30 tumors (56.6%). Tumors that were negative for both metastin and GPR54 expression were significantly larger than tumors that were positive for either metastin or GPR54 (p = 0.047). Recurrence was less frequent in patients who had metastin-positive tumors compared with those who had metastin-negative tumors (38.5% versus 70.0%, p = 0.04). Strong expression of metastin and GPR54 was significantly correlated with longer survival (p = 0.02). Metastin expression by pancreatic cancer was an independent prognostic factor for longer survival (hazard ratio, 2.1; 95% confidence interval, 1.1–4.7; p = 0.03), and the patients with a high plasma metastin level (n = 6) did not die after surgical resection.ConclusionStrong expression of metastin and GPR54 by pancreatic cancer is associated with longer survival. Metastin expression is an independent prognostic factor for the survival of pancreatic cancer patients. The plasma metastin level could become a noninvasive prognostic factor for the assessment of pancreatic cancer.


Pancreas | 2005

Analysis of E-, N-cadherin, alpha-, beta-, and gamma-catenin expression in human pancreatic carcinoma cell lines.

Eiji Toyoda; Ryuichiro Doi; Masayuki Koizumi; Kazuhiro Kami; Daisuke Ito; Tomohiko Mori; Koji Fujimoto; Sanae Nakajima; Michihiko Wada; Masayuki Imamura

Objectives: Cadherins are cell surface glycoproteins that mediate Ca2+-dependent, homophilic cell-cell adhesion. The classic cadherins interact with either β-catenin or γ-catenin, which is bound to α-catenin that links the complex to the actin cytoskeleton. It has been reported that alteration in cadherins/catenins function or expression is found in the neoplastic process as a step in metastasis. The aim of this study was to analyze the expressions of E- and N-cadherins and catenins in human pancreatic cancer cell lines. Methods: We examined the expression of cadherins and catenins in 7 human pancreatic cancer cells by RT-PCR, Western blotting, and immunocytochemistry. The interactions between cadherins and β-catenin were assessed by immunoprecipitation. Results: E-cadherin was expressed in all cell lines except for MIAPaCa-2, whereas N-cadherin was expressed in Capan-2, CFPAC-1, BxPC-3, and PANC-1. The α-, β-, and γ-catenins were expressed and cadherins/β-catenin interactions were detected in all cadherin-expressing cells. Immunocytochemical analysis showed membranous expression of cadherins and catenins. Conclusion: The decreased or loss of cadherins and catenins expression could be involved in the tumor progression and metastasis, although these events may occur in in vivo conditions by interaction between cancer cells and extracellular matrices.


Development Growth & Differentiation | 2006

Mesenchymal epimorphin is important for pancreatic duct morphogenesis

Sidhartha Tulachan; Ryuichiro Doi; Yohei Hirai; Yoshiya Kawaguchi; Masayuki Koizumi; Mark Hembree; Eri Tei; Amanda Crowley; Hooi Yew; Chris McFall; Krishna Prasadan; Barry Preuett; Masayuki Imamura; George K. Gittes

Epithelial–mesenchymal interactions are crucial for the proper development of many organs, including the pancreas. Within the pancreas, the ducts are thought to harbor stem/progenitor cells, and possibly to give rise to pancreatic ductal carcinoma. Little is known about the mechanism of formation of pancreatic ducts in the embryo. Pancreatic mesenchyme contains numerous soluble factors which help to sustain the growth and differentiation of exocrine and endocrine structures. Here, we report that one such morphoregulatory mesenchymal protein, epimorphin, plays an important role during pancreatic ductal proliferation and differentiation. We found that epimorphin is expressed in pancreatic mesenchyme during early stages of development, and at mesenchymal–epithelial interfaces surrounding the ducts at later stages. Strong upregulation of epimorphin expression was seen during in vitro pancreatic duct differentiation. Similarly, in vitro pancreatic duct formation was inhibited by a neutralizing antibody against epimorphin, whereas addition of recombinant epimorphin partially rescued duct formation. Together, our study demonstrates the role of epimorphin in pancreatic ductal morphogenesis.

Collaboration


Dive into the Masayuki Koizumi'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