Network


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

Hotspot


Dive into the research topics where Shinya Ohashi is active.

Publication


Featured researches published by Shinya Ohashi.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion

Katharine D. Grugan; Charles G. Miller; Yao Yao; Carmen Z. Michaylira; Shinya Ohashi; Andres J. Klein-Szanto; J. Alan Diehl; Meenhard Herlyn; May Han; Hiroshi Nakagawa; Anil K. Rustgi

Squamous cell cancers comprise the most common type of human epithelial cancers. One subtype, esophageal squamous cell carcinoma (ESCC), is an aggressive cancer with poor prognosis due to late diagnosis and metastasis. Factors derived from the extracellular matrix (ECM) create an environment conducive to tumor growth and invasion. Specialized cancer-associated fibroblasts (CAFs) in the ECM influence tumorigenesis. We have shown previously that the nature and activation state of fibroblasts are critical in modulating the invasive ability of ESCC in an in vivo-like organotypic 3D cell culture, a form of human tissue engineering. Dramatic differences in invasion of transformed esophageal epithelial cells depended on the type of fibroblast in the matrix. We hypothesize that CAFs create an environment primed for growth and invasion through the secretion of factors. We find that fibroblast secretion of hepatocyte growth factor (HGF) fosters the ability of transformed esophageal epithelial cells to invade into the ECM, although other unidentified factors may cooperate with HGF. Genetic modifications of both HGF in fibroblasts and its receptor Met in epithelial cells, along with pharmacologic inhibition of HGF and Met, underscore the importance of this pathway in ESCC invasion and progression. Furthermore, Met activation is increased upon combinatorial overexpression of epidermal growth factor receptor (EGFR) and p53R175H, two common genetic mutations in ESCC. These results highlight the potential benefit of the therapeutic targeting of HGF/Met signaling in ESCC and potentially other squamous cancers where this pathway is deregulated.


Cancer Research | 2010

Epidermal growth factor receptor and mutant p53 expand an esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors.

Shinya Ohashi; Mitsuteru Natsuizaka; Gabrielle S. Wong; Carmen Z. Michaylira; Katharine D. Grugan; Douglas B. Stairs; Jiri Kalabis; Maria E. Vega; Ross A. Kalman; Momo Nakagawa; Andres J. Klein-Szanto; Meenhard Herlyn; Diehl Ja; Anil K. Rustgi; Hiroshi Nakagawa

Transforming growth factor-beta (TGF-beta) is a potent inducer of epithelial to mesenchymal transition (EMT). However, it remains elusive about which molecular mechanisms determine the cellular capacity to undergo EMT in response to TGF-beta. We have found that both epidermal growth factor receptor (EGFR) overexpression and mutant p53 tumor suppressor genes contribute to the enrichment of an EMT-competent cellular subpopulation among telomerase-immortalized human esophageal epithelial cells during malignant transformation. EGFR overexpression triggers oncogene-induced senescence, accompanied by the induction of cyclin-dependent kinase inhibitors p15(INK4B), p16(INK4A), and p21. Interestingly, a subpopulation of cells emerges by negating senescence without loss of EGFR overexpression. Such cell populations express increased levels of zinc finger E-box binding (ZEB) transcription factors ZEB1 and ZEB2, and undergo EMT on TGF-beta stimulation. Enrichment of EMT-competent cells was more evident in the presence of p53 mutation, which diminished EGFR-induced senescence. RNA interference directed against ZEB resulted in the induction of p15(INK4B) and p16(INK4A), reactivating the EGFR-dependent senescence program. Importantly, TGF-beta-mediated EMT did not take place when cellular senescence programs were activated by either ZEB knockdown or the activation of wild-type p53 function. Thus, senescence checkpoint functions activated by EGFR and p53 may be evaded through the induction of ZEB, thereby allowing the expansion of an EMT-competent unique cellular subpopulation, providing novel mechanistic insights into the role of ZEB in esophageal carcinogenesis.


Pancreatology | 2005

A Case of Autoimmune Pancreatitis Associated with Sclerosing Cholangitis, Retroperitoneal Fibrosis and Sjögren’s Syndrome

Toshiro Fukui; Kazuichi Okazaki; Hazuki Yoshizawa; Shinya Ohashi; Hiroyuki Tamaki; Kimio Kawasaki; Minoru Matsuura; Masanori Asada; Hiroshi Nakase; Yasuaki Nakashima; Akiyoshi Nishio; Tsutomu Chiba

We report a very rare case of autoimmune pancreatitis (AIP) associated with sclerosing cholangitis, retroperitoneal fibrosis and Sjögren’s syndrome. The patient had an enlarged pancreas, and autoantibodies were detected in the serum. Serum IgG and IgG4 concentrations were also elevated. Endoscopic retrograde cholangiopancreatography revealed an irregular narrowing of the main pancreatic duct from the head to the body and sclerotic change in the intrapancreatic common bile duct, which later extended to the intrahepatic bile ducts. In addition, histological examination of the liver revealed lymphocytic sclerosis around the bile ducts, similar to the histology in the pancreas of AIP. Retroperitoneal tumors were diagnosed as retroperitoneal fibrosis by histological examination. Serological and functional abnormalities suggestive of Sjögren’s syndrome were detected, and histological findings of the lip were compatible with Sjögren’s syndrome. Immunohistochemistry of each lesion disclosed that most of the infiltrating lymphocytes were T cells with similar levels of both CD4+ and CD8+ cells. Moreover, some of the infiltrating plasma cells were positive for anti-IgG4 monoclonal antibody. These diseases were dramatically improved by steroid therapy. Although the pathophysiology of AIP is still unclear, the present case suggests a common pathophysiological mechanism for AIP, sclerosing cholangitis, retroperitoneal fibrosis and Sjögren’s syndrome.


Laboratory Investigation | 2005

Helicobacter felis-induced gastritis was suppressed in mice overexpressing thioredoxin-1

Kimio Kawasaki; Akiyoshi Nishio; Hajime Nakamura; Kazushige Uchida; Toshiro Fukui; Masaya Ohana; Hazuki Yoshizawa; Shinya Ohashi; Hiroyuki Tamaki; Minoru Matsuura; Masanori Asada; Toshiki Nishi; Hiroshi Nakase; Shinya Toyokuni; Wenrui Liu; Junji Yodoi; Kazuichi Okazaki; Tsutomu Chiba

Thioredoxin-1 (TRX-1) is a redox-active protein involved in scavenging reactive oxygen species and regulating redox-sensitive transcription factors. TRX-1 is induced in various inflammatory conditions and shows cytoprotective action. We investigated the roles of TRX-1 in the host defense mechanism against Helicobacter felis (H. felis) infection. Transgenic (TG) mice overexpressing human TRX-1 and wild-type (WT) mice were orally inoculated with H. felis. After 2 months, histology, oxidative damage, and gene expression of several cytokines, including macrophage inflammatory protein-2 (MIP-2), a murine equivalent to interleukin (IL)-8, in the gastric mucosa were investigated. Furthermore, the effects of TRX-1 on oxidative stress and neutrophil migration were studied both in vivo and in vitro. The gastric mucosa was thickened in H. felis-infected WT mice, but not in infected TRX-1-TG mice. Histologically, all H. felis-infected WT mice developed moderate-to-severe gastritis, whereas the development of gastritis was significantly suppressed in infected TRX-1-TG mice. Oxidative damage markers, 8-hydroxy-2′-deoxyguanosine and malondialdehyde, increased in the stomach of infected WT mice, but not TRX-1-TG mice. Upregulation of IL-1β and tumor necrosis factor-α gene expression in H. felis-infected TRX-1-TG mice was significantly lower than in WT mice. However, upregulation of MIP-2 and IL-7 was not different between the two groups. TRX-1 suppressed oxidative cytotoxicity and DNA damage, and inhibited neutrophil migration both in vivo and in vitro. The present study suggests that overexpression of TRX-1 suppresses H. felis-induced gastritis by inhibiting chemotaxis of neutrophils and reducing oxidative stress.


Cancer Biology & Therapy | 2015

EGFR inhibitors prevent induction of cancer stem-like cells in esophageal squamous cell carcinoma by suppressing epithelial-mesenchymal transition

Fumiyuki Sato; Yoshimasa Kubota; Osamu Maehara; Yutaka Hatanaka; Katsuji Marukawa; Katsumi Terashita; Goki Suda; Shunsuke Ohnishi; Yuichi Shimizu; Yoshito Komatsu; Shinya Ohashi; Shingo Kagawa; Hideaki Kinugasa; Kelly A. Whelan; Hiroshi Nakagawa; Naoya Sakamoto

There exists a highly tumorigenic subset of esophageal squamous cell carcinoma (ESCC) cells defined by high expression of CD44. A novel therapy targeting these cancer stem-like cells (CSCs) is needed to improve prognosis of ESCC. CSCs of ESCC have a mesenchymal phenotype and epithelial-mesenchymal transition (EMT) is critical to enrich and maintain CSCs. EGFR, frequently overexpressed in ESCC, has pivotal roles in EMT induced by TGF-β in invasive fronts. Thus, EMT in invasive fronts of ESCC might be important for CSCs and EGFR could be a target of a novel therapy eliminating CSCs. However, effects of EGFR inhibitors on CSCs in ESCC have not been fully examined. EGFR inhibitors, erlotinib and cetuximab, significantly suppressed enrichment of CSCs via TGF-β1-mediated EMT. Importantly, EGFR inhibitors sharply suppressed ZEB1 that is essential for EMT in ESCC. Further, EGFR inhibitors activated Notch1 and Notch3, leading to squamous cell differentiation. EGFR inhibition may suppress expression of ZEB1 and induce differentiation, thereby blocking EMT-mediated enrichment of CSCs. In organotypic 3D culture, a form of human tissue engineering, tumor cells in invasive nests showed high expression of CD44. Erlotinib significantly blocked invasion into the matrix and CD44 high expressing CSCs were markedly suppressed by erlotinib in organotypic 3D culture. In conclusion, EMT is a critical process for generation of CSCs and the invasive front of ESCC, where EMT occurs, might form a CSC niche in ESCC. EGFR inhibitors could suppress EMT in invasive fronts and be one therapeutic option targeting against generation of CSCs in ESCC.


Pancreas | 2006

Clinical significance of serum thioredoxin 1 levels in patients with acute pancreatitis

Shinya Ohashi; Akiyoshi Nishio; Hajime Nakamura; Masahiro Kido; Keiichi Kiriya; Masanori Asada; Hiroyuki Tamaki; Toshiro Fukui; Kimio Kawasaki; Norihiko Watanabe; Junji Yodoi; Kazuichi Okazaki; Tsutomu Chiba

Objective: Thioredoxin 1 (TRX-1), a redox-regulating protein with antioxidant activity, is induced by oxidative stress, and serum TRX-1 levels are recognized as an oxidative-stress marker. The aim of this study was to clarify the clinical significance of serum TRX-1 levels in patients with acute pancreatitis (AP) and evaluate the usefulness of this measurement in assessing disease severity. Methods: Serum TRX-1 levels were determined on admission in 18 patients with severe AP and 36 patients with mild AP. We also investigated the relationship between serum TRX-1 levels and clinical and laboratory data. Results: The median serum TRX-1 levels on admission were 54.9 ng/mL in mild AP and 118.8 ng/mL in severe AP. When the cutoff value for TRX-1 in predicting severe AP was determined to be 100 ng/mL, its sensitivity, specificity, and accuracy were 83.3%, 94.4%, and 90.7%, respectively. A significant correlation was observed between serum TRX-1 levels and Ranson score (r = 0.674), C-reactive protein (r = 0.718), interleukin 6 (r = 0.712), leukocyte count (r = 0.642), and serum amylase (r = 0.436). Conclusions: Serum TRX-1 levels significantly correlate with AP severity. TRX-1 should constitute a reliable oxidative-stress marker for the evaluation of AP severity in relation to oxidative stress.


The FASEB Journal | 2012

Hypoxia induces IGFBP3 in esophageal squamous cancer cells through HIF-1α-mediated mRNA transcription and continuous protein synthesis

Seiji Naganuma; Shingo Kagawa; Shinya Ohashi; Azal Ahmadi; Harry Subramanian; Sanders Chang; Kei Nakagawa; Xinjun Ji; Stephen A. Liebhaber; Andres J. Klein-Szanto; Hiroshi Nakagawa

Insulin‐like growth factor binding protein (IGFBP)‐3 regulates cell proliferation and apoptosis in esophageal squamous cell carcinoma (ESCC) cells. We have investigated how the hypoxic tumor microenvironment in ESCC fosters the induction of IGFBP3. RNA interference experiments revealed that hypoxia‐inducible factor (HIF)‐1α, but not HIF‐2α, regulates IGFBP3 mRNA induction. By chromatin immunoprecipitation and transfection assays, HIF‐1α was found to transactivate IGFBP3 through a novel hypoxia responsive element (HRE) located at 57 κb upstream from the transcription start site. Metabolic labeling experiments demonstrated hypoxia‐mediated inhibition of global protein synthesis. 7‐Methyl GTP‐cap binding assays suggested that hypoxia suppresses cap‐dependent translation. Experiments using pharmacological inhibitors for mammalian target of rapamycin (mTOR) suggested that a relatively weak mTOR activity may be sufficient for cap‐dependent translation of IGFBP3 under hypoxic conditions. Bicistronic RNA reporter transfection assays did not validate the possibility of an internal ribosome entry site as a potential mechanism for cap‐independent translation for IGFBP3 mRNA. Finally, IGFBP3 mRNA was found enriched to the polysomes. In aggregate, our study establishes IGFBP3 as a direct HIF‐1α target gene and that polysome enrichment of IGFBP3 mRNA may permit continuous translation under hypoxic conditions.—Natsuizaka, M., Naganuma, S., Kagawa, S., Ohashi, S., Ahmadi, A., Subramanian, H., Chang, S., Nakagawa, K. J., Ji, X., Liebhaber, S. A., Klein‐Szanto, A. J., Nakagawa, H. Hypoxia induces IGFBP3 in esophageal squamous cancer cells through HIF‐1α‐mediated mRNA transcription and continuous protein synthesis. FASEB J. 26, 2620‐2630 (2012). www.fasebj.org


Digestive Diseases and Sciences | 2010

Regression of B-Cell Lymphoma of the Liver with Hepatitis C Virus Infection After Treatment with Pegylated Interferon-α and Ribavirin

Yayoi Oda; Tadayuki Kou; Masaki Watanabe; Yojiro Sakuma; Nori Taguchi; Yoko Kato; Yasushi Kudo; Atsushi Yamauchi; Yasushi Sugiura; Shinya Ohashi; Masanori Asada; Toyokazu Fukunaga; Kiyotaka Kawaguchi; Hiroaki Ito; Takefumi Nakamura; Shujiro Yazumi

The pathogenesis of human B-cell lymphoma is not well understood, although some types of B-cell lymphoma seem to be related to infection with pathogens such as Helicobacter pylori [1]. Accumulating evidence reveals a high incidence of B-cell lymphoma in patients with hepatitis C virus (HCV) infection, suggesting a link between HCV infection and the development of B-cell lymphoma [2–6]. We describe here a case of primary B-cell lymphoma of the liver with HCV infection that regressed following the eradication of HCV.


Gut | 2006

Gastric mucosal hyperplasia via upregulation of gastrin induced by persistent activation of gastric innate immunity in major histocompatibility complex class II deficient mice

Toshiro Fukui; Akiyoshi Nishio; Kazuichi Okazaki; Norimitsu Uza; Satoru Ueno; Masahiro Kido; Satoko Inoue; Hiroshi Kitamura; Keiichi Kiriya; Shinya Ohashi; Masanori Asada; Hiroyuki Tamaki; Minoru Matsuura; Kimio Kawasaki; Katsumasa Suzuki; Kazushige Uchida; Hirokazu Fukui; Hiroshi Nakase; Norihiko Watanabe; Tsutomu Chiba

Background and aim: Major histocompatibility complex class II deficient (Aα0/0) mice have decreased CD4+ T cells, making them immunologically similar to patients with acquired immunodeficiency syndrome (AIDS). Both patients with AIDS and Aα0/0 mice have hypertrophic gastric folds. To clarify the mechanism of gastric mucosal hyperplasia, we investigated the pathophysiology and the role of the innate immunity in the stomach of Aα0/0 mice. Methods: Stomachs from 1–6 month old Aα0/0 mice, kept under specific pathogen free conditions, were examined at 1 month intervals histologically and immunohistochemically. Gene expression of proinflammatory cytokines, Toll-like receptors (TLRs), cyclooxygenase (COX)-2, and myeloperoxidase (MPO) activity in the gastric mucosa was investigated. Serum gastrin levels and gastric acidity were measured. Bacterial culture of the stomach was performed. To clarify the roles of hypergastrinaemia in the gastric mucosa, a gastrin receptor antagonist (AG041R) was administered. Results: Aα0/0 mice had a diffusely thick corpus mucosa with infiltration of CD11b+ granulocytes and macrophages. Anti-Ki67 staining demonstrated expansion of the proliferating neck zone. Gene expression of interleukin 1β, interferon γ, TLR-2, TLR-4, and COX-2 were upregulated, and MPO activity was increased. Only a small amount of non-pathogenic bacteria was detected in the stomach. Serum gastrin levels and Reg-Iα positive cells in the gastric mucosa increased, despite normal gastric acidity. After treatment with AG041R, gastric mucosal thickness was significantly reduced. Conclusion: Persistent activation of innate immunity in the stomach induced gastric mucosal hyperplasia through upregulation of gastrin synthesis in Aα0/0 mice, suggesting a pathophysiology similar to the gastric changes in patients with AIDS.


PLOS ONE | 2014

Preclinical Validation of Talaporfin Sodium-Mediated Photodynamic Therapy for Esophageal Squamous Cell Carcinoma

Shinya Ohashi; Osamu Kikuchi; Mihoko Tsurumaki; Yukie Nakai; Hiroi Kasai; Takahiro Horimatsu; Shin’ichi Miyamoto; Akira Shimizu; Tsutomu Chiba; Manabu Muto

Photodynamic therapy (PDT) kills cancer cells via a photochemical reaction mediated by an oncotropic photosensitizer. Herein, we performed an experimental preclinical study to validate the anti-tumour effect of talaporfin sodium-mediated PDT (t-PDT) for esophageal squamous cell carcinoma (ESCC) cells. We used human ESCC cells derived from various differentiation grades or resistant to 5-fluorouracil (5-FU). The cytotoxic effect of t-PDT was determined by evaluating cell viability, apoptosis and generation of reactive oxygen species (ROS) and DNA double-strand breaks. Furthermore, the anti-tumour effect of t-PDT was assessed using an anchorage-independent cell-growth assay and xenograft transplantation models. t-PDT induced potent cytotoxicity in ESCC cells independent of their differentiation grade or 5-FU resistance. Moreover, t-PDT induced robust apoptosis, as indicated by cell shrinkage, perinuclear vacuolization, nuclear fragmentation and induction of annexin V-positive cells. This apoptotic response was accompanied by concurrent activation of ROS, and induction of DNA double-strand breakage. Importantly, t-PDT suppressed efficiently anchorage-independent cell growth as well as ESCC-xenografted tumor formation. In aggregate, t-PDT showed anti-tumor potential for ESCC cells with various histological grades or chemoresistance, providing a novel translational rationale of t-PDT for the treatment of ESCC.

Collaboration


Dive into the Shinya Ohashi's collaboration.

Top Co-Authors

Avatar

Hiroshi Nakagawa

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshiro Fukui

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akiyoshi Nishio

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge