Network


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

Hotspot


Dive into the research topics where Toshiro Fukui is active.

Publication


Featured researches published by Toshiro Fukui.


Pancreas | 2006

Identification of a novel autoantibody against pancreatic secretory trypsin inhibitor in patients with autoimmune pancreatitis.

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

Objectives: Although autoimmune pancreatitis (AIP) has been recently recognized as a new disease entity of chronic pancreatitis, the clinical diagnosis of the disease remains disputed. Autoantibodies against carbonic anhydrase II and lactoferrin are detected in most patients with AIP, but not in about 10%. We undertook this study to determine whether additional autoantibodies are present in the serum level of AIP patients. Methods: We recruited 26 patients with AIP for the study. For comparison, we also recruited 53 patients with various pancreatic diseases and 12 healthy subjects. We immunoscreened human pancreatic cDNA library using patients sera. Positive clones were analyzed by DNA sequencing and were constructed into a pGEX-4T-1 expression vector. The recombinant proteins were used as antigens in enzyme-linked immunosorbent assay to screen the subjects sera for autoantibodies. Results: We cloned a cDNA encoding the pancreatic secretory trypsin inhibitor (PSTI). Among 26 patients with AIP, autoantibodies against PSTI were significantly positive in 11 (42.3%) by western blotting and in 8 (30.8%) by enzyme-linked immunosorbent assay, respectively. However, none of control subjects was positive for anti-PSTI antibodies. Conclusions: These findings suggest that PSTI may be related to the pathogenesis of AIP, and autoantibodies against PSTI can be a useful diagnostic marker for the disease.Abbreviations: AIP - autoimmune pancreatitis, PSTI - pancreatic secretory trypsin inhibitor, CA-II - carbonic anhydrase II, LF - lactoferrin, TBST - Tris-buffered saline containing 0.05% Tween 20, SDS-PAGE - sodium dodecyl sulfate-polyacrylamide gel electrophoresis, ELISA-enzyme-linked immunosorbent assay, OD - optical density, ANA - antinuclear antibody, AMA - antimitochondrial antibody, RF - rheumatoid factor


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.


Infection and Immunity | 2003

Involvement of myeloid dendritic cells in the development of gastric secondary lymphoid follicles in Helicobacter pylori-infected neonatally thymectomized BALB/c mice

Toshiki Nishi; Kazuichi Okazaki; Kimio Kawasaki; Toshiro Fukui; Hiroyuki Tamaki; Minoru Matsuura; Masanori Asada; Tomohiro Watanabe; Kazushige Uchida; Norihiko Watanabe; Hiroshi Nakase; Masaya Ohana; Hiroshi Hiai; Tsutomu Chiba

ABSTRACT We previously described an animal model of Helicobacter pylori-induced follicular gastritis in neonatally thymectomized (nTx) mice. However, it is still not clear whether antigen-presenting dendritic cells (DCs) in the stomach have a role in the development of secondary follicles in H. pylori-infected nTx mice. We investigated the distribution of DC subsets using this model and examined their roles. To identify lymphoid and myeloid DCs, sections were stained with anti-CD11c (pan-DC marker) in combination with anti-CD8α (lymphoid DC marker) or anti-CD11b (myeloid DC marker) and were examined with a confocal microscope. Expression of macrophage inflammatory protein 3α (MIP-3α), which chemoattracts immature DCs, was analyzed by real-time PCR and immunohistochemistry. Follicular dendritic cells (FDCs) were stained with anti-SKY28 antibodies. In noninfected nTx mice, a few myeloid and lymphoid DCs were observed in the bottom portion of the lamina propria, whereas in H. pylori-infected nTx mice, there was an increased influx of myeloid DCs throughout the lamina propria. FDC staining was also observed in the stomachs of members of the infected group. MIP-3α gene expression was upregulated in the infected nTx group, and the immunohistochemistry analysis revealed MIP-3α-positive epithelial cells. These data suggest that H. pylori infection upregulates MIP-3α gene expression in gastric epithelial cells and induces an influx of myeloid DCs in the lamina propria of the gastric mucosa in nTx mice. Myeloid DCs and FDCs might contribute to the development of gastric secondary lymphoid follicles in H. pylori-infected nTx mice.


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.


Laboratory Investigation | 2004

Immunogenetic analysis of gastric MALT lymphoma-like lesions induced by Helicobacter pylori infection in neonatally thymectomized mice

Toshiro Fukui; Kazuichi Okazaki; Hiroyuki Tamaki; Kimio Kawasaki; Minoru Matsuura; Masanori Asada; Toshiki Nishi; Kazushige Uchida; Masahiro Iwano; Masaya Ohana; Hiroshi Hiai; Tsutomu Chiba

Most gastric mucosa-associated lymphoid tissue (MALT) lymphomas are caused by Helicobacter pylori (H. pylori) infection. We previously reported that acquired lymphoid follicles with germinal centers were induced by H. pylori infection in neonatally thymectomized (nTx) mice. In the present study, we developed gastric MALT lymphoma-like lesions in nTx mice by long-term H. pylori infection, and performed immunogenetic analyses. BALB/c mice were thymectomized on the 3rd day after birth. At 6 weeks of age, mice were orally infected with 108 H. pylori and serially killed 2, 4, 6, and 12 months later. Normal BALB/c and noninfected nTx mice served as controls. Follicle formation occurred after 2 months of H. pylori infection in the nTx mice. Follicle formation and infiltration of intraepithelial lymphocytes progressed in a time-dependent manner. Lymphoepithelial lesions, a characteristic feature of MALT lymphoma, also occurred in a time-dependent manner (100% at 12 months). Serum immunoelectrophoresis revealed a monoclonal band (M-protein) in 30% (3/10) of mice 6 months after infection. M-protein-positive mice had amplification of one or two IgM and/or IgG heavy-chain genes in the gastric B lymphocytes, as determined with polymerase chain reaction, suggesting mono- or oligoclonality. Overexpression of Bcl-XL protein was immunohistologically observed in the infiltrating B lymphocytes and in some follicular B lymphocytes in 80% (8/10) of the cases at 12 months. Thus, H. pylori infection is involved in the development of gastric MALT lymphoma-like lesions in nTx mice. Our mouse model is useful for clarifying the pathogenetic mechanism of gastric MALT lymphoma by H. pylori infection.


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.


Gut | 2003

Inhibitory effects of Helicobacter pylori infection on murine autoimmune gastritis

Masaya Ohana; Kazuichi Okazaki; Chikashi Oshima; Kimio Kawasaki; Toshiro Fukui; Hiroyuki Tamaki; Minoru Matsuura; Masanori Asada; Toshiki Nishi; Kazushige Uchida; Suguru Uose; Hiroshi Nakase; Masahiro Iwano; Yumi Matsushima; Hiroshi Hiai; Tsutomu Chiba

Background and aim: Long term Helicobacter pylori infection leads to atrophic gastritis but the relation between H pylori infection and autoimmune related atrophic gastritis (AIG) remains unclear. We studied the effects of H pylori infection on the pathophysiology of AIG in mice. Materials and methods: BALB/c nu/nu mice (n=40) with or without H pylori infection received splenocytes from neonatally thymectomised mice to induce AIG. Half of the mice were orally infected with H pylori prior to AIG induction. Histological findings, and local and systemic immune responses were serially evaluated. Results: Two and six months after transfer, parietal cells in uninfected mice were depleted while those in infected mice were well preserved. The degree of gland atrophy (p<0.01), hyperplasia (p<0.01), gastric pH (p<0.05), and serum gastrin levels of infected mice were significantly lower than those of uninfected mice. Serum antiparietal cell antibody levels gradually decreased in infected mice, and were significantly lower than those of uninfected mice at six months (p<0.05). Real time polymerase chain reaction studies revealed significantly higher interleukin 4 (p<0.05) and transforming growth factor β (p<0.05) gene expression in the gastric mucosa in infected mice than in uninfected mice at both two and six months after AIG induction. Conclusions:H pylori infection inhibited the development of AIG in mice. Th2-type immune responses and transforming growth factor β in the gastric microenvironment might be involved in the inhibitory effects of H pylori infection on the development of AIG, in which Th1-type responses have an important role.


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.


Journal of Gastroenterology | 2003

Interaction of Helicobacter pylori-induced follicular gastritis and autoimmune gastritis in BALB/c mice with post-thymectomy autoimmune gastritis.

Kazuichi Okazaki; Masaya Ohana; Chikashi Oshima; Kazushige Uchida; Toshiki Nishi; Masahiro Iwano; Toshiro Fukui; Kimio Kawasaki; Minoru Matsuura; Masanori Asada; Hiroyuki Tamaki; Hiroshi Hiai; Tsutomu Chiba

BackgroundThere is still controversy about the relationship between Helicobacter pylori infection and autoimmune gastritis. The aim of this study was to clarify whether or not H. pylori infection interacts with the development of autoimmune gastritis.MethodsNeonatally thymectomized BALB/c mice with autoimmune gastritis received orally administered H. pylori and were examined histologically and serologically. The T-helper (Th)1/Th2 immune balance in the microenvironment of the stomach was evaluated by reverse-transcriptase-polymerase chain reaction (RT-PCR) for interferon (IFN)-Γ and interleukin (IL)-4.ResultsUninfected mice showed disappearance of parietal cells, and upregulation of IFN-Γ, but no germinal center formation. The infected neonatally thymectomized mice showed follicular gastritis, preserved parietal cells, decreased serum anti-parietal antibodies, and upregulation of IL-4 and IFN-Γ.ConclusionsH. pylori infection changes the microenvironment of the gastric mucosa by inducing a Th2 immune response in addition to a Th1 response, and regresses autoimmune gastritis in neonatally thymectomized BALB/c mice.


Helicobacter | 2007

Cross-primed CD8+ cytotoxic T cells induce severe Helicobacter-associated gastritis in the absence of CD4+ T cells

Toshiro Fukui; Akiyoshi Nishio; Kazuichi Okazaki; Katsuhiro Kasahara; Kazuyuki Saga; Junya Tanaka; Norimitsu Uza; Satoru Ueno; Masahiro Kido; Shinya Ohashi; Masanori Asada; Hiroshi Nakase; Norihiko Watanabe; Tsutomu Chiba

Background:u2002 Although previous studies have reported important roles of CD4+ type1‐helper T cells and regulatory T cells in Helicobacter‐associated gastritis, the significance of CD8+ cytotoxic T cells remains unknown. To study the roles of CD8+ T cells, we examined the immune response in the gastric mucosa of Helicobacter felis‐infected major histocompatibility complex (MHC) class II‐deficient (II−/–) mice, which lack CD4+ T cells.

Collaboration


Dive into the Toshiro Fukui's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge