Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 299(2): G311-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508157

ABSTRACT

Nonsteroidal anti-inflammatory drug (NSAID)-induced small intestinal injuries are serious clinical events and a successful therapeutic strategy is difficult. Regenerating gene (Reg) I protein functions as a regulator of cell proliferation and maintains intercellular integrity in the small intestine. The aim of this study was to evaluate the role of Reg I in NSAID-induced small intestinal injuries. First, to examine the effect of Reg I deficiency on such injuries, indomethacin, a widely used NSAID, was injected subcutaneously into 10-wk-old male Reg I-knockout (Reg I(-/-)) and wild-type (Reg I(+/+)) mice twice with an interval of 24 h, after which the mice were euthanized. Small intestinal injuries were assessed by gross findings, histopathology, and contents of IL-1beta and MPO in the experimental tissues. Next, we investigated the therapeutic potential of Reg I in indomethacin-induced small intestinal injuries. Recombinant Reg I protein (rReg I) was administered to 10-wk-old male ICR mice, then indomethacin was administered 6 h using the same protocol as noted above, after which small intestinal injuries were assessed after euthanasia. Our results showed that Reg I(-/-) mice had a greater number of severe small intestinal lesions after indomethacin administration. Histological examinations of the small intestines from those mice revealed deep ulcers with prominent inflammatory cell infiltration, whereas the mucosal content of proinflammatory agents was also significantly increased. In addition, rReg I administration inhibited indomethacin-induced small intestinal injuries in ICR mice. In conclusion, Reg I may be useful as a therapeutic agent in NSAID-induced small intestinal injuries.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Indomethacin/pharmacology , Intestinal Diseases/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Lithostathine/metabolism , Ulcer/pathology , Animals , Interleukin-1beta/antagonists & inhibitors , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Diseases/prevention & control , Intestine, Small/pathology , Lipopolysaccharides/pharmacology , Lithostathine/deficiency , Lithostathine/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Recombinant Proteins/pharmacology , Severity of Illness Index , Ulcer/chemically induced , Ulcer/metabolism , Ulcer/prevention & control
2.
Lab Invest ; 90(7): 1033-48, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20351696

ABSTRACT

The molecular basis of attaining columnar phenotype in Barrett's esophagus is poorly understood. One hypothesis states that factors locally produced by cells of mesenchymal origin in chronic reflux esophagitis induce metaplastic transformation. This study was performed to elucidate the factors secreted from fibroblasts that cause columnar phenotype in adjacent squamous epithelium. Human fibroblast cells were exposed to acidified medium for 20 min, followed by medium neutralization for 2 h, and then total RNA was hybridized to Sentrix Human-6 Expression BeadChips. Furthermore, esophageal mucosal biopsy specimens from reflux esophagitis patients were examined for HB-EGF expression using immunohistochemistry. In addition, cells from the human esophageal squamous epithelial cell line HET1A were treated with recombinant HB-EGF, and changes in expressions of Cdx2 and columnar markers were analyzed. The gene expression profile revealed significant upregulation of a variety of growth factors and inflammatory chemokines in response to acid exposure. Among them, HB-EGF was upregulated more than 10-fold. Biopsy specimens from reflux esophagitis patients showed a strong expression of HB-EGF in fibroblast cells underlying the damaged epithelium. Furthermore, in vitro stimulation of HET1A cells with HB-EGF increased Cdx2 in dose-dependent manners. Functional analysis of human Cdx2 promoter also revealed its upregulation by HB-EGF stimulation, showing the role of potential responsive elements (AP-1 and NF-kappaB) for its transcriptional activation. Moreover, the columnar markers cytokeratin 7 and villin were also upregulated by HB-EGF stimulation. HB-EGF induces several genes characteristics of columnar phenotypes of esophageal squamous epithelium in a paracrine manner.


Subject(s)
Barrett Esophagus/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Barrett Esophagus/pathology , CDX2 Transcription Factor , Cell Line, Tumor , Cell Survival , Cytokines/metabolism , Esophagitis, Peptic/metabolism , Esophagus/metabolism , Esophagus/pathology , Fibroblasts/pathology , Gastric Acid/physiology , Heparin-binding EGF-like Growth Factor , Humans , Metaplasia , Paracrine Communication
3.
Lab Invest ; 90(4): 556-65, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20157293

ABSTRACT

On the basis of its temporal and spatial pattern of expression during the healing of gastric ulcers, RegI is implied to be a key growth factor governing the gastric progenitor cell proliferation, which is fundamental for reconstruction of the gastric tissue; however, there is no direct in vivo evidence. The aim of this study was to use RegI-transgenic (Tg) mice to test the role of RegI protein in the healing of experimentally induced gastric ulcers. The stomachs from 48 pairs of wild-type (Wt) and Tg littermates were examined for gastric erosions after 24 h of water-immersion stress, or, 6, 12, 18 and 24 h after oral administration of HCl/ethanol. Expression levels of c-fos and c-myc proto-oncogenes were examined over time by real-time reverse transcriptase PCR to assess gastric cell proliferation. Almost all the littermate pairs tested showed superiority of Tg mice over Wt mice in the ability of decreasing ulcer index (UI) (cumulative length of erosion). The time-course study revealed that the UIs of Tg were lower in the healing phase, and not in the injury phase. The fraction of proliferating cells was higher in Tg mice than in Wt mice throughout the time course as assessed by c-fos expression levels. This is the first in vivo evidence that RegI has a role in gastric ulcer healing. We suggest that RegI exerts its effects by promoting growth and not by cytoprotection.


Subject(s)
Gastric Mucosa/physiopathology , Lithostathine/physiology , Wound Healing/physiology , Animals , Disease Models, Animal , Female , Gastric Mucosa/metabolism , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Stomach Ulcer
4.
J Immunol ; 182(11): 7222-32, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454719

ABSTRACT

MFG-E8 (milk fat globule-epidermal growth factor 8) deficiency is strongly associated with acquisition of immune-mediated disorders due to the loss of tissue homeostasis. However, comparatively little is known regarding its functions in gastrointestinal tract disorders, in which immune homeostasis is a major concern. Herein, we report altered MFG-E8 expression in inflamed colons during the acute phase of murine experimental colitis and found that treatment with recombinant MFG-E8, but not its arginine-glycine-aspartate mutant counterpart, ameliorated colitis by reducing inflammation and improving disease parameters. To reveal the MFG-E8-mediated antiinflammatory mechanism, we employed an in vitro system, which showed the down-regulation of NF-kappaB in an LPS-dependent manner. Additionally, MFG-E8 altered alpha(v)beta(3) integrin-mediated focal adhesion kinase phosphorylation by impeding the binding of one of its potent ligands osteopontin, which becomes activated during colitis. Taken together, our results indicated that MFG-E8 has a novel therapeutic potential for treatment of colitis.


Subject(s)
Antigens, Surface/pharmacology , Inflammation , Integrin alphaVbeta3/metabolism , Intestines/pathology , Milk Proteins/pharmacology , Osteopontin/metabolism , Animals , Antigens, Surface/analysis , Colitis , Disease Models, Animal , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Milk Proteins/analysis , NF-kappa B/analysis , Phosphorylation , Signal Transduction/drug effects
5.
Lab Invest ; 88(6): 652-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18458670

ABSTRACT

Various therapies are used for inflammatory bowel diseases (IBD), though none seem to be extremely effective. AP-1 is a major transcription factor that upregulates genes involved in immune and proinflammatory responses. We investigated decoy oligodeoxynucleotide (ODN) targeting AP-1 to prevent dextran sulfate sodium (DSS)-induced colitis in mice. Functional efficacies of synthetic decoy and scrambled ODNs were evaluated in vitro by a reporter gene luciferase assay and measuring flagellin-induced IL-8 expression by HCT-15 cells transfected with ODNs. Experimental colitis was induced in mice with a 2.5% DSS solution in drinking water for 7 days, and decoy or scrambled ODNs were intraperitoneally injected from days 2 to 5. Colitis was assessed by weight loss, colon length, histopathology, and detection of myeloperoxidase (MPO), IL-1beta, and TNF-alpha in colon tissue. Therapeutic effects of AP-1 and NF-kappaB decoy ODNs were compared. Transfection of AP-1 decoy ODN inhibited AP-1 transcriptional activity in reporter assays and flagellin-induced IL-8 production in vitro. In mice, AP-1 decoy ODN, but not scrambled ODN, significantly inhibited weight loss, colon shortening, and histological inflammation induced by DSS. Further, AP-1 decoy ODN decreased MPO, IL-1beta, and TNF-alpha in colonic tissue of mice with DSS-induced colitis. The AP-1 decoy therapeutic effect was comparable to that of NF-kappaB decoy ODN, which also significantly decreased intestinal inflammation. Double-strand decoy ODN targeting AP-1 effectively attenuated intestinal inflammation associated with experimental colitis in mice, indicating the potential of targeting proinflammatory transcription factors in new therapies for IBD.


Subject(s)
Colitis, Ulcerative/metabolism , Inflammation/drug therapy , Oligonucleotides/pharmacology , Transcription Factor AP-1/metabolism , Animals , Cell Culture Techniques , Cell Line, Tumor , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colonic Neoplasms/pathology , Dextran Sulfate/pharmacology , Flow Cytometry , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , Genes, Reporter , Humans , Interleukin-8/antagonists & inhibitors , Luciferases/metabolism , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Specific Pathogen-Free Organisms , Transcription, Genetic/drug effects , Transfection
6.
Apoptosis ; 13(5): 609-20, 2008 May.
Article in English | MEDLINE | ID: mdl-18392683

ABSTRACT

The lactogenic hormone prolactin (PRL) regulates milk protein gene expression in mammary glands. To maintain homeostatic balance in the body, milk fat globule epidermal growth factor 8 (MFG-E8) is vital for phagocytic clearance of apoptotic cells. We investigated the effects of PRL on MFG-E8 expression in macrophages by evaluating its promoter function. Macrophages were stimulated with PRL, and the expression of MFG-E8 was determined using real-time PCR and Western blotting. The role of MFG-E8 on phagocytosis of apoptotic cells in PRL-treated macrophages was assessed using microscopy, while the response of PRL to MFG-E8 expression was evaluated using luciferase assay. Following treatment with PRL, significant up-regulations of the PRL receptor and MFG-E8 were observed in macrophages, though PRL-treated macrophages more efficiently engulfed apoptotic cells. The results of MFG-E8 promoter analysis showed considerable up-regulation of promoter activity in macrophages following PRL treatment and results from mutation analysis of the MFG-E8 promoter suggested that the C/EBPbeta binding site was responsible for PRL-induced activation of the MFG-E8 promoter. C/EBPbeta activity was found to be up-regulated in PRL-treated cells as revealed by an electrophoretic mobility shift assay (EMSA). In conclusion, PRL is a potent inducer of MFG-E8 expression in macrophages, while its effect is mediated by the presence of a responsive element in the MFG-E8 promoter.


Subject(s)
Antigens, Surface/biosynthesis , Apoptosis/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Milk Proteins/biosynthesis , Prolactin/pharmacology , Animals , Antigens, Surface/genetics , Base Sequence , Cell Line , Cell Nucleus/metabolism , Mice , Milk Proteins/genetics , Phagocytosis/drug effects , Promoter Regions, Genetic , Protein Transport , Receptors, Prolactin/biosynthesis , Up-Regulation
7.
J Gastroenterol Hepatol ; 23(2): 296-302, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18289358

ABSTRACT

BACKGROUND AND AIM: Accelerated cellular proliferation in Barrett's esophagus has been implicated in Barrett's elongation and malignant transformation. Therefore, growth factors may play important roles in the pathophysiology of Barrett's esophagus. Regenerating gene (REG), an epithelial growth factor, has been reported to link mucosal inflammation and subsequent carcinogenesis in the gastrointestinal tract. The aim of this study was to investigate whether REG is expressed in Barrett's esophagus and to elucidate the relationship between REG protein expression and clinicopathological factors of Barrett's esophagus. METHODS: Between July 2003 and June 2004, 266 patients with endoscopically and histologically proven Barrett's esophagus were enrolled in this study. Before endoscopic examination, all participants were requested to answer structured questionnaires on gastroesophageal reflux symptoms and drugs usage. Mucin phenotype, cyclooxygenase-2 expression, cellular proliferation, apoptosis and REG Ialpha protein expression were investigated in the biopsy samples taken from Barrett's esophagus. Clinicopathological factors that correlated with REG Ialpha protein expression in patients with Barrett's esophagus were evaluated using multivariate logistic regression analysis. RESULTS: REG Ialpha protein expression was observed in 48 (18.0%) of 266 patients with Barrett's esophagus by immunohistochemistry. Newly developed squamous re-epithelialization of Barrett's esophagus at biopsy sites, presence of hiatal hernia and aging were shown to correlate with REG Ialpha protein expression. CONCLUSIONS: The present study is the first to show REG expression in Barrett's esophagus. Expression of REG Ialpha was more frequently observed in patients who showed squamous re-epithelialization of Barrett's esophagus at biopsy sites.


Subject(s)
Barrett Esophagus/metabolism , Lithostathine/metabolism , Aged , Aging/metabolism , Barrett Esophagus/complications , Barrett Esophagus/pathology , Biopsy , Esophagitis, Peptic/complications , Female , Helicobacter Infections/complications , Helicobacter pylori , Hernia, Hiatal/complications , Humans , Immunohistochemistry , Male , Middle Aged , Predictive Value of Tests , Transforming Growth Factor alpha/metabolism , Transforming Growth Factor beta/metabolism
8.
Curr Pharm Des ; 12(32): 4215-28, 2006.
Article in English | MEDLINE | ID: mdl-17100624

ABSTRACT

Toll-like receptors (TLRs) are sensors of microbial products that initiate host defense responses in multicellular organisms. They are mainly linked to innate immunity and bridging to adaptive immunity, signaling through different TLRs responsible for a wide range of biological responses. The intracellular signaling pathways through Toll/interleukin-1 receptor (IL-1R) domains result in recruitment of the cytoplasmic adaptor molecules, with subsequent activation of a signaling cascade leading to nuclear factor-kappa B (NF-kappaB). TLR-signaling induces host inflammatory response and the inflammation becomes more severe in the absence of several extra and intra cellular negative regulators of TLR-signaling. In the intestine, TLR-dependent activation of NF-kappaB plays a vital role in maintaining epithelial homeostasis as well as regulating infections and inflammation, while dysregulation of TLR-signaling is associated with the pathogenesis of inflammatory bowel diseases (IBD). Recent findings regarding innate immunity-mediated regulation of intestinal pathophysiology prove that development of new drugs targeting TLRs including antagonists of TLR-signaling and agonists of their negative regulators has a potential impact on therapeutic strategies for intestinal inflammatory diseases.


Subject(s)
Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/immunology , Inflammation/drug therapy , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology , Animals , Gastrointestinal Diseases/pathology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Signal Transduction
9.
Am J Physiol Gastrointest Liver Physiol ; 291(4): G735-43, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16959957

ABSTRACT

Midkine (MK) is a unique growth and differentiation factor that modulates the proliferation and migration of various cells; however, little is known regarding its relationship to intestinal diseases. The aim of this study was to investigate MK expression and its role in dextran sulfate sodium (DSS)-induced colitis in rats. The expressions of MK, receptor-like protein-tyrosine phosphatase (RPTP)-beta, and proinflammatory cytokines were examined in rat colonic tissues after the development of DSS-induced colitis using Northern blotting, immunohistochemistry, and laser-capture microdissection (LCM) coupled with RT-PCR. The effects of MK on the migration of intestinal epithelial cells (IEC-6) were also evaluated in vitro using an intestinal wound repair model. MK expression was significantly increased in damaged colonic mucosa, mainly from day 3 to day 5 after the end of DSS administration, with abundant MK immunoreactive signals detected in submucosal fibroblasts. Expressions of proinflammatory cytokines were most strongly induced on day 1, which preceded the augmentation of MK expression. Results of LCM coupled with RT-PCR clearly indicated RPTP-beta expression in colonic epithelial cells. The migration assay showed that wound repair in the MK-treated groups was accelerated dose dependently. The present results showed for the first time that intestinal inflammation upregulates the MK-RPTP-beta system, which may stimulate mucosal regeneration during the process of healing of colitis. Additional investigations regarding the role of MK may contribute to the development of new options for the treatment of inflammatory bowel diseases.


Subject(s)
Colitis/metabolism , Colon/metabolism , Cytokines/metabolism , Gene Expression Regulation , Wound Healing , Animals , Cell Line , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Colitis/chemically induced , Colitis/pathology , Cytokines/genetics , Dextran Sulfate/pharmacology , Fibroblasts/metabolism , Interleukin-1/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Midkine , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
J Gastroenterol ; 41(7): 706-10, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16933010

ABSTRACT

Eosinophilic esophagitis (EE) is a rarely diagnosed condition involving eosinophilic infiltration of the esophageal mucosa. Here we present a case of EE in a 69-year-old Japanese man, who presented with abdominal pain, appetite loss, and a history of bronchial asthma. Laboratory findings included peripheral eosinophilia and an increased serum immunoglobulin E level. Computed tomography showed diffuse severe thickening of the esophageal wall, and a barium esophagogram revealed a small caliber of the middle and lower portion of the esophagus, without normal peristaltic contractions. Endoscopy of the esophagus showed a pale mucosa, with adherent whitish exudates resembling fungal infection, and prominent ring-like contractions. Histologic examination of a biopsy specimen revealed marked eosinophil infiltration into the esophageal mucosa. Endoscopic ultrasonography (EUS) demonstrated marked circumferential thickening of the esophageal submucosal layer, and an esophageal manometry study showed a high percentage of ineffective esophageal peristalsis and high-amplitude esophageal body contractions. EUS findings showed no change even after oral corticosteroid therapy, although the histological findings were improved. This is thought to be the first documented Japanese case of EE. EE should be considered in the differential diagnosis in cases of esophageal motility disturbance, even if the patients do not complain of dysphagia.


Subject(s)
Esophagitis , Aged , Eosinophilia/diagnosis , Eosinophilia/diagnostic imaging , Eosinophilia/immunology , Esophagitis/diagnosis , Esophagitis/diagnostic imaging , Esophagitis/immunology , Esophagus/diagnostic imaging , Humans , Japan , Male , Radiography , Ultrasonography
11.
Clin Vaccine Immunol ; 13(1): 132-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16426010

ABSTRACT

We recently demonstrated that the pattern recognition receptors (PRRs) toll-like receptor 2 (TLR2), TLR4, and CD14 are expressed in mouse colonic epithelium in a compartmentalized manner. Here we report the localization of TLR5, the receptor for bacterial flagellin, and its distinctive down-regulation during experimental colitis. Guts from normal BALB/c mice and those with dextran sodium sulfate (DSS)-induced colitis were compared. Each gut was divided into seven segments (stomach, small intestine [three parts], and colon [three parts]), and epithelial cells and crypt units were collected by scraping and EDTA treatment, respectively. Northern blotting showed that TLR5 mRNA was preferentially expressed in the epithelium of the proximal colon in normal mice. Laser capture microdissection coupled to reverse transcriptase PCR confirmed this localization. TLR5 protein expression reflected mRNA expression, as evidenced by Western blotting. In mice with acute colitis, inflammation occurred mainly in the distal colon. Interestingly, while TLR2, TLR4, and CD14 were up-regulated in the inflamed colon, TLR5 was down-regulated at both the mRNA and protein levels. Decreased TLR5 expression was more evident during chronic colitis. Additional in vitro studies using a mouse cell line, Colon-26, showed that gamma interferon (IFN-gamma) time- and dose-dependently down-regulates TLR5. In conclusion, epithelial cells, mainly in the proximal colon, constitutively express TLR5. TLR5 expression is down-regulated in vivo during acute and chronic DSS-induced colitis, in contrast to the expression of TLR2, TLR4, and CD14. The mechanism governing TLR5 regulation may therefore differ from that controlling other PRRs. Finally, IFN-gamma may be involved in down-regulating TLR5 expression.


Subject(s)
Cecum/chemistry , Colitis/metabolism , Down-Regulation , Toll-Like Receptor 5/metabolism , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/immunology , Disease Models, Animal , Disease Progression , Epithelium , Humans , Interferon-gamma/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mice , RNA, Messenger/biosynthesis , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/immunology , Up-Regulation
12.
Biochem Biophys Res Commun ; 339(2): 540-7, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16300730

ABSTRACT

RNA polymerase III promoters of human ribonuclease P RNA component H1, human U6, and mouse U6 small nuclear RNA genes are commonly used in short hairpin RNA (shRNA) expression vectors due their precise initiation and termination sites. During transient transfection of shRNA vectors, we observed that H1 or U6 promoters also express longer transcripts enough to express several reporter genes including firefly luciferase, green fluorescent protein EGFP, and red fluorescent protein JRed. Expression of such longer transcripts was augmented by upstream RNA polymerase II enhancers and completely inhibited by downstream polyA signal sequences. Moreover, the transcription of firefly luciferase from human H1 promoter was sensitive to RNA polymerase II inhibitor alpha-amanitin. Our findings suggest that commonly used polymerase III promoters in shRNA vectors are also prone to RNA polymerase II mediated transcription, which may have negative impacts on their targeted use.


Subject(s)
Genetic Vectors/genetics , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , RNA Polymerase III/metabolism , RNA Polymerase II/metabolism , RNA/genetics , Transcription, Genetic/genetics , Animals , Cell Line , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Genes, Reporter/genetics , Genetic Vectors/chemistry , Humans , Mice , RNA/chemistry , RNA Polymerase II/antagonists & inhibitors
13.
Am J Gastroenterol ; 100(10): 2157-66, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16181363

ABSTRACT

BACKGROUND & AIM: Chronic inflammation induced by Helicobacter pylori infection is closely associated with epithelial cell proliferation and apoptosis, which are related to cellular turnover in gastric mucosa. Reg protein is a regenerating gene product and a potent growth factor for gastric mucosal cells, however, little is known regarding its association with the pathogenesis of H. pylori infection. The aim of this study was to investigate Reg protein production and its regulation in H. pylori-associated gastritis. METHODS: Gastric fundic biopsy samples were taken from patients with and without H. pylori infection. In vivo expression of Reg protein was examined by Western blotting and immunohistochemistry methods. The effects of interleukin (IL)-8 on Reg protein expression and transcriptional activation of the Reg gene in ECC10 cells were investigated by Western blotting and luciferase assays, respectively. RESULTS: Reg expression was found localized in the deeper part of gastric fundic glands and clearly shown in chromogranin A-positive cells in the gastric corpus. Semiquantitative immunohistochemistry and Western blotting results for Reg expression were significantly associated with polymorphonuclear neutrophil activity and chronic inflammation of gastric mucosa. IL-8 production in the gastric mucosa was significantly augmented by H. pylori infection, while IL-8 dose-dependently stimulated Reg protein production and Reg promoter activity in vitro in cultured ECC10 cells. CONCLUSION: The present study showed for the first time that Reg protein may be a potent stimulator of gastric epithelial cells in H. pylori-infected human gastric mucosa stimulated by IL-8. Further, our findings provide evidence of a novel link between Reg protein and H. pylori infection, which may help explain the molecular mechanisms underlying H. pylori-associated diseases, including gastric cancer.


Subject(s)
Calcium-Binding Proteins/metabolism , Gastric Mucosa/metabolism , Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Interleukin-8/physiology , Nerve Tissue Proteins/metabolism , Adult , Aged , Cell Culture Techniques , Female , Gastritis/microbiology , Humans , Lithostathine , Male , Middle Aged , Neutrophil Infiltration , Neutrophils/physiology
14.
Genes Cells ; 9(11): 1113-23, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15507122

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) acts as a ligand-activated transcription factor. Although ligand-induced cellular differentiation and growth inhibition have been mostly studied on human cancers expressing PPARgamma, it is unclear if the transcriptional activation of PPARgamma is the main mechanism of growth inhibition. In this study, we investigated whether there is a link between growth inhibitory effect and transcriptional activation of PPARgamma in several gastrointestinal tumour cell lines. The transcriptional activation potential of PPARgamma was assessed by reporter gene assay employing a PPRE-luciferase vector, and growth inhibitory effect of PPARgamma was investigated by (3)H-thymidine incorporation assay, in the presence or absence of thiazolidinedione ligands, rosiglitazone and troglitazone. As expected, in the case of cell lines positive for the transcriptional activation potential of PPARgamma (T.Tn, MKN-45 and LoVo), both the ligands induced growth inhibition. However, in case of some other cell lines negative for the transcriptional activation potential of PPARgamma (TT, AGS and HCT-15), troglitazone still showed a growth inhibitory effect. Administration of the PPARgamma antagonist GW9662 did not reverse this growth inhibitory activity of troglitazone. The introduction of dominant negative mutants of PPARgamma did not suppress the activity either. These observations suggest that while rosiglitazone inhibits cellular growth predominantly through transcriptional activation of PPARgamma, troglitazone can induce it both in PPARgamma-dependent and -independent pathways.


Subject(s)
Cell Division/physiology , Gastrointestinal Neoplasms/pathology , PPAR gamma/physiology , Base Sequence , Cell Line, Tumor , DNA Primers , DNA, Complementary , Enzyme Activation , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Thiazolidinediones/pharmacology , Transcriptional Activation , Transfection
15.
J Gastroenterol ; 39(6): 507-13, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15235866

ABSTRACT

In 1984, Reg protein was shown to be stimulated during the regeneration of pancreatic islets. Since then, many Reg-related proteins have been identified in humans and other animals. These Reg-related proteins are classified into four subfamilies according to their amino-acid sequences, but they share a similar structure and physiological function. The role of Reg in gastric tissue was investigated, and Reg I was found to be expressed mainly in gastric fundic enterochromaffin-like (ECL) cells. Reg I production in ECL cells is stimulated by gastrin, as well as by the proinflammatory cytokine, cytokine-induced neutrophil chemoattractant (CINC)-2Beta. In patients with chronic hypergastrinemia, Reg production is stimulated, with the increased proliferation of gastric mucosal cells. Patients with Helicobacter pylori infection also showed increased Reg production in the gastric mucosa, partly via increased plasma gastrin concentration and partly via increased proinflammatory cytokine production. Thus, Reg protein induced by H. pylori infection may be partly responsible for the increased proliferation of gastric epithelial cells in H. pylori-infected patients. Reg protein is also produced in many gastric cancer cells, especially in poorly differentiated and advanced cancers. Reg protein stimulates the proliferation of several gastric cancer cell types, and gastric cancers with Reg protein expression tend to show a poorer clinical outcome. In summary, Reg protein may be a growth factor that regulates the proliferation and differentiation of normal and neoplastic gastric epithelial cells.


Subject(s)
Gastric Mucosa/cytology , Animals , Cell Division , Chronic Disease , Enterochromaffin-like Cells/metabolism , Gastric Mucosa/metabolism , Gastrins/physiology , Gastritis/metabolism , Helicobacter Infections/physiopathology , Helicobacter pylori/cytology , Humans , Immunohistochemistry , In Situ Hybridization , Stomach Neoplasms/physiopathology
16.
J Immunol ; 173(2): 1406-16, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15240737

ABSTRACT

TLR4, a member of pattern recognition receptors, is the main receptor of LPS. MD-2 physically associates with TLR4 on the cell surface and confers LPS responsiveness. Helicobacter pylori LPS is one of the major virulence factors for induction of gastritis. We demonstrated in this study the role of MD-2 in TLR4-dependent signaling in H. pylori-associated gastritis. Gastric biopsy samples collected from patients with and without H. pylori infection and four gastric cancer cell lines were used for this study. TLR-4 and MD-2 expression in biopsy specimens and the cell lines was examined by using RT-PCR. Localization of TLR-4 in histological sections was evaluated by immunohistochemistry. For in vitro functional assays, we established stable transfectants of AGS cells expressing TLR4 and MD-2. Cellular distribution of TLR4 was examined by flow cytometry. NF-kappaB activation and activation of IL-8 and MD-2 promoters were assessed by reporter gene assay. H. pylori infection up-regulated the TLR4 and MD-2 expression in gastric mucosa. TLR4 staining was observed predominantly in epithelial cells, located in both the cytoplasm and at the apical surface. MD-2 transfection in AGS cells markedly increased cell surface expression of TLR4 and augmented the activation of NF-kappaB and IL-8 promoter upon stimulation with H. pylori LPS. Live H. pylori also stimulated transcriptional activation of MD-2. This study revealed that MD-2 expression is elevated in gastric epithelial cells during H. pylori infection, suggesting that the TLR4/MD-2 system is a potent receptor complex involved in the response to H. pylori LPS in the stomach.


Subject(s)
Antigens, Surface/metabolism , Gastritis/metabolism , Helicobacter Infections/immunology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Adult , Antigens, Surface/genetics , Female , Gastritis/immunology , Gastritis/microbiology , Genes, Reporter , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Humans , Interleukin-8/genetics , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96 , Male , Membrane Glycoproteins/genetics , Middle Aged , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Toll-Like Receptor 4 , Toll-Like Receptors
17.
Oncogene ; 23(20): 3572-9, 2004 Apr 29.
Article in English | MEDLINE | ID: mdl-15116092

ABSTRACT

Reg (regenerating gene product) was originally identified as a growth factor involved in pancreatic regeneration. During the healing course of gastric erosion, Reg expression is highly increased in the enterochromaffin-like (ECL) cells surrounding the ulcer crater, suggesting its role as a regulator of gastric mucosal regeneration. However, there has been no direct in vivo evidence of a growth-promoting role of Reg for the gastric mucosal cells. In the current study, Reg-transgenic mice were created and gastric mucosa were analysed for histological changes. Transgenic mice showed a marked increase in the thickness of the fundic mucosa. Anti-proliferating cell nuclear antigen (PCNA) staining of the fundic mucosa demonstrated the enlargement of the proliferating neck zone and the lower PCNA-negative zone. Histological analysis employing antibodies against cell-type markers revealed expansion of the chief cell and parietal cell populations and no change in the number of surface mucus-producing cells, ECL cells, or G cells. In conclusion, Reg has a growth-promoting effect on gastric progenitor cells and an activity to direct the differentiation of the cells into chief cell and parietal cell lineages. This was in contrast to other factors, all of which had been shown to drive differentiation towards mucus producing cells in vivo. In the injured gastric mucosa, Reg may play a unique and important part in the reconstruction of the properly organized mucosal architecture.


Subject(s)
Calcium-Binding Proteins/genetics , Cell Differentiation/physiology , Chief Cells, Gastric/physiology , Glycoproteins/genetics , Parietal Cells, Gastric/physiology , Animals , Apoptosis/physiology , Calcium-Binding Proteins/metabolism , Cell Division/physiology , Gastric Mucosa/cytology , Gastric Mucosa/growth & development , Gastric Mucosa/pathology , Glycoproteins/metabolism , Lithostathine , Mice , Mice, Transgenic
18.
Am J Physiol Gastrointest Liver Physiol ; 286(3): G508-14, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14563668

ABSTRACT

For the production and vesicle storage of histamine, Enterochromaffin-like (ECL) cells express histidine decarboxylase (HDC) and vesicular monoamine transporter 2 (VMAT2). Although HDC and VMAT2 show dynamic changes during gastric ulcer healing, the control system of their expression has not been fully investigated. In the present study, we investigated the effect of transforming growth factor-alpha (TGF-alpha) and proinflammatory cytokines on HDC and VMAT2 expression in rat ECL cells. Time course changes in the expression of TGF-alpha during the healing of acetic acid-induced ulcers were studied. EGF receptor (EGFR) expression was also examined in ECL cells, whereas the direct effects of TGF-alpha and proinflammatory cytokines on HDC and VMAT2 expression in ECL cells were investigated using in vivo and in vitro models. During the process of ulcer healing, expression of TGF-alpha mRNA was markedly augmented. Furthermore, EGFR was identified in isolated ECL cells. TGF-alpha stimulated HDC and VMAT2 mRNA expression and protein production and also increased histamine release from ECL cells. Selective EGFR tyrosine kinase inhibitor tyrphostin AG1478 almost completely inhibited HDC and VMAT2 gene expression induced by TGF-alpha in vivo and in vitro. During gastric mucosal injury, TGF-alpha was found to stimulate ECL cell functions by increasing HDC and VMAT2 expression.


Subject(s)
Enterochromaffin Cells/metabolism , Histidine Decarboxylase/biosynthesis , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins , Neuropeptides , Transforming Growth Factor alpha/pharmacology , Acetic Acid , Animals , Blotting, Northern , Cell Separation , Cells, Cultured , Enterochromaffin Cells/drug effects , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Gene Expression Regulation, Enzymologic/drug effects , Histidine Decarboxylase/metabolism , Immunohistochemistry , Kinetics , Male , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
19.
Cancer Lett ; 199(2): 169-73, 2003 Sep 25.
Article in English | MEDLINE | ID: mdl-12969789

ABSTRACT

We investigated the frequency of BRAF mutations in human pancreatic cancer specimens to determine its role in the development of pancreatic cancer. Nine pancreatic cancer samples without a K-ras codon 12 mutation and 19 with a K-ras mutation were included in the study. Analyses of the BRAF sequence revealed mutations in exon 15 (V599E) in two cases, both of which also exhibited a K-ras codon 12 mutation. No BRAF mutation was found in cases without a K-ras mutation. The BRAF V599E mutation was not found to be a major mutation in pancreatic cancers that had no K-ras codon 12 mutation.


Subject(s)
Genes, ras/genetics , Mutation/genetics , Oncogene Proteins/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Adenocarcinoma, Mucinous/etiology , Adenocarcinoma, Mucinous/genetics , Aged , Aged, 80 and over , Carcinoma, Papillary/etiology , Carcinoma, Papillary/genetics , DNA Mutational Analysis , DNA Primers/chemistry , Female , Humans , Male , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/etiology , Polymerase Chain Reaction , Proto-Oncogene Proteins B-raf , Survival Rate , Tumor Cells, Cultured
20.
J Immunol ; 170(8): 3977-85, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12682225

ABSTRACT

Pattern recognition receptors (PRRs), which include the Toll-like receptors (TLRs), are involved in the innate immune response to infection. TLR4 is a model for the TLR family and is the main LPS receptor. We wanted to determine the expression of TLR4 and compare it with that of TLR2 and CD14 along the gastrointestinal mucosa of normal and colitic BALB/c mice. Colitis was induced with 2.5% dextran sodium sulfate (DSS). Mucosa from seven segments of the digestive tract (stomach, small intestine in three parts, and colon in three parts) was isolated by two different methods. Mucosal TLR4, CD14, TLR2, MyD88, and IL-1beta mRNA were semiquantified by Northern blotting. TLR4 protein was determined by Western blotting. TLR4/MD-2 complex and CD14 were evaluated by immunohistochemistry. PRR genes were constitutively expressed and were especially stronger in colon. TLR4 and CD14 mRNA were increased in the distal colon, but TLR2 mRNA was expressed more strongly in the proximal colon, and MyD88 had a uniform expression throughout the gut. Accordingly, TLR4 and CD14 protein levels were higher in the distal colon. TLR4/MD-2 and CD14 were localized at crypt bottom epithelial cells. TLR4/MD2, but not CD14, was found in mucosal mononuclear cells. Finally, DSS-induced inflammation was localized in the distal colon. All genes studied were up-regulated during DSS-induced inflammation, but the normal colon-stressed gut distribution was preserved. Our findings demonstrate that TLR4, CD14, and TLR2 are expressed in a compartmentalized manner in the mouse gut and provide novel information about the in vivo localization of PRRs.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Antigens, Ly/biosynthesis , Antigens, Ly/genetics , Antigens, Ly/metabolism , Blotting, Northern , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Regulation/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lymphocyte Antigen 96 , Macromolecular Substances , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Models, Animal , Myeloid Differentiation Factor 88 , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...