Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Mucosal Immunol ; 12(4): 980-989, 2019 07.
Article in English | MEDLINE | ID: mdl-31076664

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces cell apoptosis by transducing apoptosis signals. Recently, accumulating evidence demonstrated that TRAIL regulates autoimmune inflammation and immune cell homeostasis in several autoimmune animal models, suggesting a novel immunoregulatory role of TRAIL in autoimmune diseases. However, the impact of TRAIL in inflammatory bowel disease is yet undefined. This study is to address the therapeutic effects and immunoregulatory role of TRAIL in autoimmune gut inflammation. We demonstrated herein that TRAIL significantly suppressed gut inflammation and reduced the severity of colitis in a dextran sodium sulfate (DSS)-induced colitis model. Suppression of gut inflammation was not due to induction of apoptosis in colonic T cells, dendritic cells, or epithelium cells by TRAIL. In contrast, TRAIL directly inhibited activation of colitogenic T cells and development of gut inflammation in an adoptive transfer-induced colitis model. The anti-inflammatory effects of TRAIL on colitis were abolished when T cells from TRAIL receptor (TRAIL-R) knockout mice were adoptively transferred, suggesting that TRAIL regulates autoreactive colitogenic T-cell activation in the development of gut inflammation. Our results demonstrate that TRAIL effectively inhibited colonic T-cell activation and suppressed autoimmune colitis, suggesting a potential therapeutic application of TRAIL in human inflammatory bowel disease.


Subject(s)
Colitis/etiology , Colitis/metabolism , Lymphocyte Activation/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Autoimmunity , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout
2.
Scand J Rheumatol ; 45(4): 304-11, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26771445

ABSTRACT

OBJECTIVES: This phase IIIB study compared the efficacy and safety of febuxostat and allopurinol in gout patients with or without tophi who were HLA-B*5801 negative. METHOD: Eligible patients were randomized to a febuxostat group (80 mg QD) or an allopurinol group (300 mg QD). Following an initial 2-week washout period, over the next 12 weeks we made five measurements of serum urate levels along with assessments of adverse events (AEs). RESULTS: Forty-three out of 152 screened subjects (28.3%) were ineligible either because of the presence of the HLA-B*5801 allele or for various other reasons. The febuxostat group (n = 54) and the allopurinol group (n = 55) had no significant differences in demographic or baseline characteristics. From week 2 to week 12, the febuxostat group had a significantly lower serum urate level than the allopurinol group (p ≤ 0.001 for all comparisons) and significantly more patients with serum urate levels less than 6.0 mg/dL. The serum urate levels of the febuxostat group declined by more than 40% from week 2 to week 12 and this decrease was greater than that in the allopurinol group (~30%). The two groups were similar in terms of AEs. CONCLUSIONS: Febuxostat was more effective than allopurinol in reducing the serum urate levels of Han Chinese patients with gout or tophaceous gout who were HLA-B*5801 negative, without causing any serious skin reactions. Febuxostat should be considered for treatment of Han Chinese patients with gout who are HLA-B*5801 negative.


Subject(s)
Febuxostat/therapeutic use , Gout Suppressants/therapeutic use , Gout/drug therapy , Hyperuricemia/drug therapy , Administration, Oral , Adult , Alleles , Allopurinol/therapeutic use , China , Female , Gout/blood , Gout/genetics , HLA-B Antigens/genetics , Humans , Hyperuricemia/blood , Male , Middle Aged , Treatment Outcome , Uric Acid/blood
3.
Blood Cancer J ; 4: eXX, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25303370

ABSTRACT

Chronic Helicobacter pylori-stimulated immune reactions determine the pathogenesis of gastric mucosa-associated lymphoid tissue (MALT) lymphoma. We aimed to explore the genetic predisposition to this lymphoma and its clinical implication. A total of 68 patients and 140 unrelated controls were genotyped for 84 single-nucleotide polymorphisms in genes encoding cytokines, chemokines and related receptors that play important roles in T cell-mediated gastrointestinal immunity. Five genotypes in IL-22, namely CC at rs1179246, CC at rs2227485, AA at rs4913428, AA at rs1026788 and TT at rs7314777, were associated with disease susceptibility. The former four genotypes resided in the same linkage disequilibrium block (r(2)=0.99) that conferred an approximately threefold higher risk. In vitro experiments demonstrated that co-culturing peripheral mononuclear cells or CD4(+) T cells with H. pylori stimulated the secretion of interleukin-22 (IL-22), and that IL-22 induced the expression of antimicrobial proteins, RegIIIα and lipocalin-2, in gastric epithelial cells. Furthermore, patients with gastric tissue expressing IL-22 were more likely to respond to H. pylori eradication (14/22 vs 4/19, P<0.006). We conclude that susceptibility of gastric MALT lymphoma is influenced by genetic polymorphisms in IL-22, the product of which is involved in mucosal immunity against H. pylori and associated with tumor response to H. pylori eradication.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Helicobacter Infections , Helicobacter pylori , Interleukins , Lymphoma, B-Cell, Marginal Zone , Neoplasm Proteins , Polymorphism, Single Nucleotide , Stomach Neoplasms , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/therapy , Humans , Interleukins/biosynthesis , Interleukins/genetics , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, B-Cell, Marginal Zone/microbiology , Lymphoma, B-Cell, Marginal Zone/therapy , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/therapy , Interleukin-22
4.
Blood Cancer J ; 4: e220, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24949857

ABSTRACT

We recently showed that Helicobacter pylori (HP)-positive gastric 'pure' diffuse large B-cell lymphoma (DLBCL) may respond to HP eradication therapy. However, whether these HP-related 'pure' DLBCL of the stomach may differ fundamentally from those unrelated to HP remains unclear. In this study, we compared the clinicopathologic features of these two groups of patients who had been uniformly treated by conventional chemotherapy. Forty-six patients were designated HP-positive and 49 were HP-negative by conventional criteria. HP-positive patients had a lower International Prognostic Index score (0-1, 65% vs 43%, P=0.029), a lower clinical stage (I-IIE1, 70% vs 39%, P=0.003), a better tumor response to chemotherapy (complete pathologic response, 76% vs 47%, P=0.004) and significantly superior 5-year event-free survival (EFS) (71.7% vs 31.8%, P<0.001) and overall survival (OS) (76.1% vs 39.8%, P<0.001). To draw a closer biologic link with HP, HP-positive tumors were further examined for CagA expression in lymphoma cells. Compared with CagA-negative cases (n=16), CagA-positive cases (n=27) were associated with high phosphorylated SHP-2 expression (P=0.016), and even better 5-year EFS (85.2% vs 46.3%, P=0.002) and OS (88.9% vs 52.9%, P=0.003). HP-related gastric 'pure' DLBCL may be a distinct tumor entity, which is less aggressive, and responds better to conventional chemotherapy.


Subject(s)
Helicobacter Infections/physiopathology , Helicobacter pylori/isolation & purification , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/microbiology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology , Antigens, Bacterial/biosynthesis , Bacterial Proteins/biosynthesis , Disease-Free Survival , Female , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Prognosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
5.
Cell Death Dis ; 5: e1109, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24603337

ABSTRACT

Helicobacter pylori (H. pylori) infection is associated with chronic gastritis, peptic ulcer and gastric cancer. Apoptosis induced by microbial infections is implicated in the pathogenesis of H. pylori infection. Here we show that human gastric epithelial cells sensitized to H. pylori confer susceptibility to TRAIL-mediated apoptosis via modulation of death receptor signaling. Human gastric epithelial cells are intrinsically resistant to TRAIL-mediated apoptosis. The induction of TRAIL sensitivity by H. pylori is dependent on the activation of caspase-8 and its downstream pathway. H. pylori induces caspase-8 activation via enhanced assembly of the TRAIL death-inducing signaling complex (DISC) through downregulation of cellular FLICE-inhibitory protein (FLIP). Overexpression of FLIP abolished the H. pylori-induced TRAIL sensitivity in human gastric epithelial cells. Our study thus demonstrates that H. pylori induces sensitivity to TRAIL apoptosis by regulation of FLIP and assembly of DISC, which initiates caspase activation, resulting in the breakdown of resistance to apoptosis, and provides insight into the pathogenesis of gastric damage in Helicobacter infection. Modulation of host apoptosis signaling by bacterial interaction adds a new dimension to the pathogenesis of Helicobacter.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Epithelial Cells/drug effects , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Stomach/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Stomach/microbiology , Stomach/pathology , Time Factors
8.
Blood Cancer J ; 3: e125, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23852160

ABSTRACT

We previously reported that CagA can be translocated into B cells in Helicobacter pylori (HP) coculture media, and the translocation appears biologically significant as activation of the relevant cellular pathways was noticed. In this study, we further explore if CagA can be detected in malignant B cells of HP-positive gastric mucosa-associated lymphoid tissue (MALT) lymphoma. Expression of CagA was evaluated by immunohistochemistry. CagA expression was further confirmed by western blot analysis. The association between CagA expression in malignant B cells and tumor response to HP eradication therapy (HPE) was evaluated in 64 stage IE gastric MALT lymphoma patients. We detected CagA expression in 31 (48.4%) of 64 patients: 26 (68.4%) of the 38 HP-dependent cases and 5 (19.2%) of the 26 HP-independent cases (P<0.001). Patients with CagA expression responded to HPE quicker than those without (median time to complete remission, 3.0 vs 6.5 months, P=0.025). Our results indicated that CagA can be translocated into malignant B cells of MALT lymphoma, and the translocation is clinically and biologically significant.

9.
Clin Exp Immunol ; 161(3): 551-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20646001

ABSTRACT

B7-H1 [programmed death-ligand-1 (PD-L1)] is a B7-family member that binds to programmed death-1 (PD-1). Recently, deficiency of PD-L1 has been demonstrated to result in accelerated gastric epithelial cell damage in gastritis, and PD-L1 is suggested to play a critical role in regulating T cell homeostasis. Here, we aimed to gain more insight into gastric PD-L1 expression, regulation and function during Helicobacter pylori infection. PD-L1 expression in human gastric epithelial cells was analysed using Western blotting, quantitative polymerase chain reaction and fluorescence activated cell sorter analysis. Furthermore, co-culture experiments of human gastric epithelial cells with primary human T cells or Jurkat T cells were conducted. PD-L1 expression in primary human gastric epithelial cells was strongly enhanced by H. pylori infection and activated T cells, and augmented markedly by further stimulation with interferon-γ or tumour necrosis factor-α. Moreover, PD-L1 expression in gastric epithelial cells significantly induced apoptosis of T cells. Our results indicate that a novel bidirectional interaction between human gastric epithelial cells and lymphocytes modulates PD-L1 expression in human gastric epithelial cells, contributing to the unique immunological properties of the stomach.


Subject(s)
Antigens, CD/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression/genetics , Helicobacter pylori/physiology , Antigens, CD/metabolism , Apoptosis/drug effects , B7-H1 Antigen , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Flow Cytometry , Gene Expression/drug effects , Host-Pathogen Interactions , Humans , Interferon-gamma/pharmacology , Jurkat Cells , Lymphocyte Activation , Pyloric Antrum/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/pharmacology
10.
Lupus ; 18(8): 676-81, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19502262

ABSTRACT

Milk fat globule-EGF factor 8 (MFG-E8) is a molecule implicated in phagocytic clearance of apoptotic cells by bridging between macrophages and apoptotic cells. Defects in MFG-E8 cause lupus-like disease in murine models. The aim of our study is to determine whether genetic variation in MFG-E8 predisposes human to systemic lupus erythematosus (SLE). A case-control study of MFG-E8 genetic polymorphism was performed on 147 SLE patients and 146 non-lupus control subjects. Single nucleotide polymorphisms (SNPs) in the coding sequence of human MFG-E8 gene were investigated. SNPs on MFG-E8 residues 3 (3(Arg or Ser)) and 76 (76(Leu or Met)) did not show genetic linkage. Genetic polymorphism on MFG-E8 residue 76 correlated significantly to SLE. The MFG-E8-76(Met) allele predisposed subjects to SLE in a recessive mode (odds ratio: 2.1, P = 0.020), while carriage of MFG-E8-76(Leu) were negatively associated with SLE. The MFG-E8 genotypic combinations with 3(Ser) and 76(Leu) showed the most pronounced protective effect on SLE when compared to the most predisposing genotype 3(Arg/Arg)-76(Met/Met) (OR: 0.29, P = 0.007). According to our result, MFG-E8 is associated with SLE predisposition in Taiwanese. Our study implicates that the impairment of phagocytic clearance of apoptotic cells through phosphotidylserine-dependent MFG-E8 system may lead to the development of human SLE.


Subject(s)
Antigens, Surface/genetics , Genetic Predisposition to Disease , Lupus Erythematosus, Systemic/genetics , Milk Proteins/genetics , Polymorphism, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , Case-Control Studies , DNA Mutational Analysis , Female , Genotype , Humans , Male , Middle Aged , Molecular Sequence Data , Phagocytes/metabolism , Young Adult
11.
Clin Exp Immunol ; 151(3): 383-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18190609

ABSTRACT

Decoy receptor 3 (DcR3/TR6) is a decoy receptor for the Fas ligand (FasL) and can inhibit FasL-induced apoptosis. It has been reported recently that DcR3 can induce T cell activation via co-stimulation of T cells, suggesting that DcR3 may be involved in the pathophysiology of autoimmune diseases. This study aims to analyse the serum DcR3 in patients with systemic lupus erythematosus (SLE) and to investigate the role of DcR3 in the pathogenesis of SLE. Significantly elevated serum DcR3 was observed in SLE patients, and the mean serum DcR3 level was significantly higher for those with active disease [SLE disease activity index (SLEDAI) >/= 10] compared with that in patients with inactive disease (SLEDAI < 10). In addition to reducing activation-induced cell death in activated T cells via neutralization of the FasL, soluble DcR3-Fc enhanced T cell proliferation and increased interleukin-2 and interferon-gamma production via co-stimulation of T cells. Moreover, enhanced T cell reactivity to DcR3-induced co-stimulation was demonstrated in lymphocytes from patients with SLE, suggesting the elevated serum DcR3 may associate with enhanced T cell activation in vivo. These findings are the first to demonstrate that serum DcR3 concentrations are increased in SLE patients, and this may imply a possible role of DcR3 in the pathogenesis of SLE via enhanced T cell hyperreactivity and reduced apoptosis in activated T cells.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Member 6b/blood , T-Lymphocytes/immunology , Adult , Apoptosis/immunology , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Middle Aged , Severity of Illness Index
13.
Aliment Pharmacol Ther ; 20(2): 203-11, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15233701

ABSTRACT

BACKGROUND: The host genetic factors that determine the clinical outcomes of Helicobacter pylori-infected individuals remain unclear. AIM: To elucidate the risks of host interleukin-1 (IL-1) genetic polymorphisms and H. pylori infection in the development of gastric cancer. METHODS: In a case-control study of 164 controls and 142 patients with gastric cancer, the IL-1B-511 biallelic polymorphisms and the IL-1RN penta-allelic variable number of tandem repeats were genotyped. RESULTS: The carriage of IL-1RN*2, male gender, old age and H. pylori infection independently increased the risk of gastric cancer, with odds ratios of 3.3 [95% confidence interval (CI), 1.4-7.7], 2.1 (95% CI, 1.2-3.8), 5.3 (95% CI, 3.1-9.0) and 2.2 (95% CI, 1.3-3.8), respectively. H. pylori-infected individuals who were carriers of IL-1RN*2 showed increased risks of both intestinal and diffuse types of gastric cancer, with odds ratios of 11.0 and 8.7, respectively. In addition, these individuals also had a higher score of intestinal metaplasia in the corpus than did uninfected non-carriers. CONCLUSIONS: This study is the first to verify IL-1RN*2 as an independent factor governing the development of gastric cancer in Asian individuals. A combination of H. pylori testing and host genotyping may target the eradication of H. pylori to high-risk individuals.


Subject(s)
Adenocarcinoma/genetics , Helicobacter Infections/genetics , Helicobacter pylori/genetics , Interleukin-1/genetics , Polymorphism, Genetic/genetics , Stomach Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Male , Middle Aged , Risk Factors , Stomach Neoplasms/microbiology
14.
Curr Med Res Opin ; 19(4): 336-41, 2003.
Article in English | MEDLINE | ID: mdl-12841927

ABSTRACT

A multi-centre, double-blind, randomised parallel-group study was conducted to compare the efficacy and safety between etodolac SR and diclofenac in treating patients with osteoarthritis of the knee. Thirty-two patients receiving etodolac SR (400 mg/day) and 32 patients receiving diclofenac (100 mg/day) were included for analyses. After receiving the study treatment, etodolac SR and diclofenac were shown to produce comparable improvement in pain relief as measured by a 10-cm visual analogue scale. There was no statistically significant difference observed between groups for the degree of functional impairment or the amount of paracetamol taken. The etodolac SR treatment group showed fewer incidents of adverse event. In addition, fewer gastrointestinal symptoms were observed in the etodolac SR treatment group. A statistically higher percentage of etodolac SR-treated patients were better tolerated to the study treatment compared with the diclofenac-treated patients. Etodolac SR-treated patients also took fewer antacids than the diclofenac-treated patients during the treatment period. In conclusion, etodolac SR is an effective treatment with fewer side-effects than diclofenac for patients with osteoarthritis of the knee.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Diclofenac/therapeutic use , Etodolac/therapeutic use , Knee Joint , Osteoarthritis/drug therapy , Abdominal Pain/etiology , Aged , Analysis of Variance , Delayed-Action Preparations , Diclofenac/adverse effects , Double-Blind Method , Etodolac/adverse effects , Female , Gastrointestinal Hemorrhage/etiology , Humans , Male , Middle Aged
16.
Gut ; 51(1): 15-20, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12077085

ABSTRACT

BACKGROUND AND AIMS: A subset of non-ulcer dyspepsia (NUD) disorders can evolve into peptic ulcer disease. This prospective study attempted to determine the independent risk factors for ulcer formation in NUD patients, and compared the natural history of Helicobacter pylori positive and negative NUD subjects. METHODS: From May 1997 to April 1999, consecutive NUD patients were enrolled into the study. Endoscopy was performed routinely on enrolment, at the end of the second and 12th months, and whenever there was a dyspepsia attack. Patients were prospectively followed up for two years. RESULTS: Peptic ulcers occurred in 16 of 209 NUD patients during the two year follow up period. Multivariate analysis of 13 host and bacterial factors demonstrated that advanced age (odds ratio 2.90), H pylori infection (odds ratio 3.59), and use of non-steroidal anti-inflammatory drugs (NSAID; odds ratio 4.46) were independently significant in predicting subsequent peptic ulcer development. NUD patients with all three risk factors had a 75% (3/4) risk of developing peptic ulcer but the ulcer incidence in patients without any of the risk parameters was only 1.2% (1/84). The resolution rate of symptoms in the H pylori positive NUD patients was similar to the H pylori negative patients (57.9% v 49.1%; 95% confidence interval (CI) -5 to 22). However, rates for subsequent peptic ulcer and erosion development were significantly higher in H pylori positive patients than in H pylori negative patients (ulcer 12.6% v 3.5%, 95% CI 1-16; erosion 23.2% v 12.3%, 95% CI 1-21). CONCLUSION: A small but significant proportion of NUD patients develop peptic ulcer after long term follow up. H pylori infection, NSAID use, and advanced age are independent risk factors for subsequent ulcer formation. Follow up endoscopy is strongly indicated for an NUD patient with multiple risk factors for ulcer formation when symptoms recur.


Subject(s)
Dyspepsia/complications , Peptic Ulcer/etiology , Age Factors , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Chi-Square Distribution , Dyspepsia/drug therapy , Dyspepsia/microbiology , Female , Follow-Up Studies , Helicobacter Infections/complications , Helicobacter pylori , Humans , Incidence , Male , Middle Aged , Peptic Ulcer/epidemiology , Prospective Studies , Regression Analysis , Risk Factors
17.
J Immunol ; 167(3): 1347-52, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466352

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL, also called Apo2L), a novel member of TNF superfamily, induces apoptosis in transformed cell lines of diverse origin. TRAIL is expressed in most of the cells, and the expression is up-regulated in activated T cells. Four receptors for TRAIL have been identified, and there is complex interplay between TRAIL and TRAIL receptors in vivo. The actual biological function of TRAIL/TRAIL receptor is still not clear. Growing evidence has demonstrated that members of TNF superfamily transduce signals after engagement with their receptors. Cross-linking of TRAIL by plate-bound rTRAIL receptor, death receptor 4-Fc fusion protein enhanced T cell proliferation and increased IFN-gamma production in conjunction with immobilized suboptimal anti-CD3 stimulation in mouse splenocytes. The increase of T cell proliferation by death receptor 4-Fc was dose dependent, and this effect could be blocked by soluble rTRAIL proteins, indicating the occurrence of reverse signaling through TRAIL on T cell. The enhanced secretion of IFN-gamma mediated via TRAIL could be blocked by SB203580, a p38 mitogen-activated protein kinase-specific inhibitor. Thus, in addition to its role in inducing apoptosis by binding to the death receptors, TRAIL itself can enhance T cell proliferation after TCR engagement and signal the augmentation of IFN-gamma secretion via a p38-dependent pathway. This provides another example of reverse signaling by a member of TNF superfamily. In conclusion, our data suggest that TRAIL can itself transduce a reverse signal, and this may shed light on the biological function of TRAIL.


Subject(s)
Apoptosis/immunology , Interferon-gamma/biosynthesis , Lymphocyte Activation , Membrane Glycoproteins/physiology , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis/genetics , Apoptosis Regulatory Proteins , CD3 Complex/immunology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Ligands , Lymphocyte Activation/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyridines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , T-Lymphocytes/enzymology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases
18.
J Biomed Sci ; 8(3): 262-9, 2001.
Article in English | MEDLINE | ID: mdl-11385298

ABSTRACT

Fas (CD95) and Fas ligand (FasL/CD95L) are involved in programmed cell death and the regulation of host immune responses. FasL has been shown to provide immune privilege, thus prolonging the survival of unmatched grafts in a variety of tissues, such as eyes and testis. In murine FasL (mFasL) transgenic mice, FasL provoked granulocyte infiltration and insulitis in the pancreas. We intended to study whether the expression of human FasL, instead of mFasL, on mouse beta islet cells could avoid granulocyte infiltration, and whether islet cells transgenic for FasL could be used in islet transplantation. We produced transgenic mice in which the human FasL transgene was driven by rat insulin promoter and was expressed exclusively in the pancreas islet cells in ICR mice. In contrast to mFasL transgenic mice, histochemical staining showed that the pancreas was intact in human FasL transgenic ICR mice. However, when human FasL transgenic islet cells were transplanted into allogeneic mice with streptozotocin-induced diabetes, human FasL appeared not to prolong graft survival. Intensive granulocyte infiltration into the islet grafts was observed in recipients (Balb/c mice) which received islet grafts from human FasL transgenic mice, but not from nontransgenic, allogeneic ICR mice on day 31. Our observations suggest that FasL alone is insufficient to confer immune protection, and that other environmental factors might contribute to the formation of immune privilege sites in vivo


Subject(s)
Graft Survival/immunology , Immune Tolerance/immunology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Membrane Glycoproteins/metabolism , Animals , Cell Death , Diabetes Mellitus, Experimental/immunology , Fas Ligand Protein , Humans , Inflammation/pathology , Insulin/genetics , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/pathology , Jurkat Cells , Membrane Glycoproteins/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Neutrophil Infiltration , Promoter Regions, Genetic/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Transgenes/genetics
19.
J Immunol ; 166(5): 3309-14, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207286

ABSTRACT

Mouse mammary tumor viruses encode superantigens that interact with MHC class II proteins and stimulate T cells. We show here that presentation of mouse mammary tumor virus superantigen does not require DM. Furthermore, we have identified a strong class II peptide binding motif in the Mtv-7 superantigen, and we show that this motif is necessary for association with class II molecules in in vitro translation and in vivo functional assays. Our results suggest that endogenously synthesized viral superantigen can bind to MHC class II heterodimers during biosynthesis in the endoplasmic reticulum in a manner analogous to that used by the class II-associated invariant chain.


Subject(s)
HLA-D Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Mammary Tumor Virus, Mouse/immunology , Superantigens/immunology , Superantigens/metabolism , Amino Acid Motifs , Animals , Antigen Presentation/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Viral , Cell-Free System/immunology , Dimerization , Dogs , HLA-DR1 Antigen/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Models, Immunological , Mutation , Oligopeptides/immunology , Oligopeptides/metabolism , Protein Binding/immunology , Protein Biosynthesis/immunology , Rabbits , Superantigens/biosynthesis , Superantigens/genetics
20.
Aliment Pharmacol Ther ; 15(2): 195-201, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11148437

ABSTRACT

BACKGROUND: Although the eradication of Helicobacter pylori infection benefits patients with gastric or duodenal ulcers, the value of eradicating the infection in the patients with functional dyspepsia (FD) remains controversial. AIMS: To determine whether eradicating H. pylori can prevent the subsequent development of ulcers or relieve the symptoms of functional dyspepsia patients. METHODS: In a double-blind, placebo-controlled trial, 161 patients infected with H. pylori who had functional dyspepsia were randomly assigned to 7 days of treatment with a lansoprazole-based triple therapy or placebo and then followed for 1 year. The main outcome measures were the development of peptic ulcers and the resolution of symptoms. RESULTS: H. pylori was eradicated in 63 out of 81 patients (78%) in the treatment group and none of the 80 patients (0%) in the placebo group. During the follow-up period, two patients in the treatment group and six patients in the placebo group developed peptic ulcers at repeat endoscopy (2.5% vs. 7.5%; 95% CI: -12 to 2). The reduction in ulcer rates was statistically significant in the 'ulcer-like' sub-group (0% vs. 16.7%; 95% CI: -32 to -2), but not in the 'dysmotility-like' and 'unclassifiable' sub-groups. Regarding symptom response, the resolution rates of symptoms were similar between the treatment and placebo groups (58.0% vs. 55.0%, 95% CI: -12 to 18). Additionally, no significant differences existed in the symptom responses between the treatment and control arms in each of the dyspepsia sub-groups. CONCLUSIONS: Eradicating H. pylori can prevent the subsequent development of peptic ulcers in the patients with 'ulcer-like' functional dyspepsia. However, this approach does not significantly reduce the symptoms of functional dyspepsia patients.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Dyspepsia/drug therapy , Helicobacter Infections/drug therapy , Helicobacter pylori , Omeprazole/analogs & derivatives , Omeprazole/therapeutic use , Peptic Ulcer/prevention & control , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Aged , Aged, 80 and over , Double-Blind Method , Drug Therapy, Combination , Dyspepsia/complications , Dyspepsia/microbiology , Female , Helicobacter Infections/complications , Helicobacter pylori/drug effects , Humans , Lansoprazole , Male , Middle Aged , Peptic Ulcer/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...