Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Clin Exp Immunol ; 169(2): 129-36, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22774987

ABSTRACT

It has been reported that interferon (IFN)-γ-secreting T cells reactive to gluten can be detected in the peripheral blood of individuals with treated coeliac disease (CD) after a short consumption of wheat-containing food. By contrast, very little is known about the reproducibility of this in-vivo procedure in the same patient cohort which underwent two, or more, gluten consumptions. Fourteen coeliac patients in remission consumed wheat bread for 3 days; 13 underwent a second gluten challenge after a wash-out of 3-10 months on a strict gluten-free diet. Immune reactivity to gluten was analysed in peripheral blood by detecting IFN-γ before and 6 days after commencing a gluten diet. Gliadin-specific IFN-γ-secreting CD4(+) T cells increased significantly on day 6 of the first challenge. These cells resulted as prevalently human leucocyte antigen (HLA)-DQ restricted and with a phenotype of gut homing, as suggested by the expression of ß7-integrin. Similarly, reactiveness to gliadin was observed after the second wheat consumption, although with an individual variability of responses at each challenge. Our findings confirmed that the short wheat challenge is a non-invasive approach to investigate the gluten-related immune response in peripheral blood of subjects intolerant to gluten. Furthermore, we demonstrated that the in-vivo procedure can be reproduced in the same subject cohort after a gluten wash-out of at least 3 months. Our study has important implications for the application of this procedure to clinical practice.


Subject(s)
Antigens, Plant/immunology , Celiac Disease/diagnosis , Celiac Disease/immunology , Glutens/immunology , Triticum/immunology , Adolescent , Adult , Antigens, Plant/administration & dosage , Epitopes/chemistry , Epitopes/immunology , Female , Gliadin/chemistry , Gliadin/immunology , HLA-DQ Antigens/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Peptides/chemistry , Peptides/immunology , Time Factors , Triticum/chemistry , Young Adult
2.
Scand J Immunol ; 74(4): 335-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21615450

ABSTRACT

Coeliac disease (CD) is a very common food-sensitive enteropathy, which is triggered by gluten ingestion and is mediated by CD4(+) T cells. In addition, alterations in the intestinal microbiota that is normally involved in the homeostasis of GALT (gut-associated lymphoid tissue) seem to play a role in CD. In accordance with these findings, we previously reported that Lactobacillus casei can induce a strong enhancement of the T cell-mediated response to gliadin without inducing enteropathy. In this study, we analysed the effects of L. casei administration in a mouse model of gliadin-induced villous damage that was recently developed and involves the inhibition of cyclo-oxygenase (COX) activities in gliadin-sensitized HLA-DQ8 transgenic mice. To address the issue, we assessed the weight loss, the intestinal cytokine pattern, the density of CD25(+) cells and morphometry of the gut mucosa. We confirmed that COX inhibition in sensitized mice caused villus blunting, dysregulated expression of tumour necrosis factor (TNF)-α and reduced gliadin-specific IL-2 production. Notably, the administration of probiotic strain induced a complete recovery of villus blunting. This finding was associated with a delay in weight decrease and a recovery of basal TNF-α levels, whereas the numbers of CD25(+) cells and the levels of IL-2 remained unchanged. In conclusion, our data suggest that the administration of L. casei can be effective in rescuing the normal mucosal architecture and GALT homeostasis in a mouse model of gliadin-induced enteropathy.


Subject(s)
Celiac Disease , Gliadin/immunology , HLA-DQ Antigens , Lacticaseibacillus casei/immunology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/pathology , Cytokines/analysis , Gliadin/metabolism , Glutens , Immunologic Factors , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Weight Loss
3.
Thorax ; 60(9): 773-80, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15994249

ABSTRACT

BACKGROUND: Cystic fibrosis (CF) airways are characterised by chronic inflammation, increased interleukin (IL)-8 secretion, and neutrophil activation which are considered the principal factors of morbidity and mortality in CF patients. Optimising management of this chronic inflammatory response is therefore a key issue of basic and clinical CF research. Several reports have addressed ways to manage CF airways inflammation, and an attractive therapeutic strategy may be the inhibition of the p38-mitogen activated protein kinase (p38-MAP-k) pathway. METHODS: A new ex vivo model was used to study the mucosal inflammatory response to environmental airways stimuli. Nasal biopsy tissues from CF patients and controls were cultured ex vivo for 20 minutes, 4 hours, and 24 hours in the presence of lipopolysaccharide (LPS) from Pseudomonas aeruginosa (PA) with and without the p38-MAP-k inhibitor SB203580. Quantitative mRNA assessment, immunohistochemistry, and Western blots were used to detect the expression and modulation of inflammatory markers. RESULTS: PA-LPS challenge induced a time dependent mucosal inflammation indicated by rapid epithelial activation, IL-8 release, COX-2 upregulation, and neutrophil migration to the upper mucosal layers. Some of these LPS induced changes (IL-8 release and neutrophil migration) were specific to CF tissues. SB203580 significantly controlled all LPS induced mucosal changes in CF tissues. CONCLUSION: These findings provide a rationale and proof of principle for the potential use of p38-MAP-k inhibitors to control inflammation in patients with CF.


Subject(s)
Bronchitis/enzymology , Cystic Fibrosis/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adolescent , Adult , Blotting, Western , Bronchitis/prevention & control , Cells, Cultured , Cyclooxygenase 2 , Cystic Fibrosis/pathology , Cystic Fibrosis/prevention & control , Female , Humans , Interleukin-8/analysis , Lipopolysaccharides/pharmacology , Male , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/analysis , Pseudomonas aeruginosa , RNA, Messenger/analysis , Respiratory Mucosa
4.
Diabetologia ; 48(5): 931-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15830185

ABSTRACT

AIMS/HYPOTHESIS: A deranged mucosal immune response and dietary factors may play an important role in the pathogenesis of type 1 diabetes. The aims of our work were to look for the presence of small intestinal enteropathy in non-obese diabetic (NOD) mice in relation to the presence of wheat proteins in the diet, and to assess their role in the risk of developing diabetes. METHODS: Female NOD mice were fed a standard or gluten-free diet or a gluten-free diet with the addition of wheat proteins (MGFD). Small intestine architecture, intraepithelial CD3(+) infiltration, epithelial expression of H2-IA, mRNA for IFN-gamma and IL-4 were assessed. RESULTS: NOD mice fed a standard diet showed reduced villous height, increased intraepithelial infiltration by CD3(+) cells and enhanced expression of H2-IA and IFN-gamma mRNA when compared with mice on the gluten-free diet. The cumulative diabetes incidence at 43 weeks of age was 65% in the latter and 97% in the former (p<0.01). Mice on MGFD also showed increased epithelial infiltration and a higher incidence of diabetes. CONCLUSIONS/INTERPRETATION: Mice fed a wheat-containing diet showed a higher incidence of diabetes, signs of small intestinal enteropathy and higher mucosal levels of proinflammatory cytokines.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diet , Intestinal Diseases/etiology , Triticum/adverse effects , Animal Feed , Animals , Disease Models, Animal , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , Jejunum/pathology , Mice , Mice, Inbred NOD
5.
Gut ; 54(1): 46-53, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15591503

ABSTRACT

BACKGROUND: Enteropathy in coeliac disease (CD) is sustained by a gliadin specific Th1 response. Interleukin (IL)-10 can downregulate Th1 immune responses. AIM: We investigated the ability of recombinant human (rh) IL-10 to suppress gliadin induced Th1 response. PATIENTS AND METHODS: IL-10 RNA transcripts were analysed by competitive reverse transcription-polymerase chain reaction in duodenal biopsies from untreated and treated CD patients, non-coeliac enteropathies (NCE), and controls. CD biopsies were cultured with a peptic-tryptic digest of gliadin with or without rhIL-10. The proportion of CD80+ and CD25+ cells in the lamina propria, epithelial expression of Fas, intraepithelial infiltration of CD3+ cells, as well as cytokine synthesis (interferon gamma (IFN-gamma) and IL-2) were measured. Short term T cell lines (TCLs) obtained from treated CD biopsies cultured with gliadin with or without rhIL-10 were analysed by ELISPOT for gliadin specific production of IFN-gamma. RESULTS: In untreated CD and NCE, IL-10 RNA transcripts were significantly upregulated. In ex vivo organ cultures, rhIL-10 downregulated gliadin induced cytokine synthesis, inhibited intraepithelial migration of CD3+ cells, and reduced the proportion of lamina propria CD25+ and CD80+ cells whereas it did not interfere with epithelial Fas expression. In short term TCLs, rhIL-10 abrogated the IFN-gamma response to gliadin. CONCLUSIONS: rhIL-10 suppresses gliadin specific T cell activation. It may interfere with the antigen presenting capacity of lamina propria mononuclear cells as it reduces the expression of CD80. Interestingly, rhIL-10 also induces a long term hyporesponsiveness of gliadin specific mucosal T cells. These results offer new perspectives for therapeutic strategies in coeliac patients based on immune modulation by IL-10.


Subject(s)
Celiac Disease/immunology , Gliadin/immunology , Immune Tolerance , Interleukin-10/immunology , Th1 Cells/immunology , Adolescent , Adult , Celiac Disease/diet therapy , Cell Line , Child , Child, Preschool , Gene Expression Regulation/immunology , Humans , Infant , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Middle Aged , Organ Culture Techniques , RNA, Messenger/genetics , Recombinant Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , T-Lymphocyte Subsets/immunology
6.
Amino Acids ; 26(4): 381-6, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15290344

ABSTRACT

Celiac disease (CD) is an autoimmune pathology of the small intestine triggered, in genetically predisposed patients, by the exposition to gliadin, a flour protein, thus evoking local immune reactions and mucosal atrophy. The discovery that type 2 transglutaminase (TG2) is the main, if not the sole, target of the endomysium CD-specific autoantibodies assigned to this enzyme a master regulator role of CD. Two separated events, both based on the finding that gliadin is able to act as a TG2 substrate, have been described to indicate that TG2 is involved in both the humoral and cellular immune responses. In this paper we review the novel insights on the localization and enzymatic activity of TG2 in the small intestinal mucosa. Moreover, we report on the capability of gliadin and its peptides to act as TG2 substrates.


Subject(s)
Celiac Disease/enzymology , GTP-Binding Proteins/metabolism , Gliadin/metabolism , Transglutaminases/metabolism , Glutens/chemistry , Glutens/genetics , Glutens/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Intestine, Small/anatomy & histology , Intestine, Small/metabolism , Isoenzymes/metabolism , Peptides/genetics , Peptides/metabolism , Protein Glutamine gamma Glutamyltransferase 2
7.
Gut ; 52(1): 57-62, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12477760

ABSTRACT

BACKGROUND: Studies on intestinal T cell clones from the mucosa of patients with coeliac disease have led to the identification of immunogenic gliadin epitopes. One is HLA-DQ8 restricted, its recognition by T cells being increased by introduction of negatively charged residues operated by tissue transglutaminase. AIM: To test HLA-DQ8 restricted epitope in both native (QYPSGQGSFQPSQQNPQA) and deamidated (QYPSGEGSFQPSQENPQA) forms in an organ culture system of treated coeliac mucosa from HLA-DQ8 positive and HLA-DQ8 negative patients. PATIENTS AND METHODS: Jejunal biopsies obtained from 10 patients with coeliac disease (six HLA-DQ8 positive and four HLA-DQ8 negative) were cultured in vitro with a peptic-tryptic digest (PT) of gliadin, or with the native (peptide A) or deamidated (peptide B) peptide. Intraepithelial CD3(+) and lamina propria total CD25(+) and CD3(+)CD25(+) cells were counted, lamina propria intercellular adhesion molecule 1 (ICAM-1) expression was evaluated, as well as that of Fas molecules on epithelial cells. RESULTS: In HLA-DQ8 positive, but not in HLA-DQ8 negative, coeliacs the density of intraepithelial CD3(+) cells, lamina propria total CD25(+), and CD3(+)CD25(+) cells, as well as expression of ICAM-1 and Fas molecules were significantly increased in biopsies cultured with PT, peptide A, or peptide B compared with biopsies cultured in medium alone. CONCLUSION: These data show that the DQ8 restricted gliadin peptide is immunogenic only in the intestinal mucosa of HLA-DQ8 positive coeliac patients in both native and deamidated forms.


Subject(s)
Celiac Disease/immunology , Epitopes/immunology , Gliadin/immunology , HLA-DQ Antigens/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Adolescent , Adult , CD3 Complex , Female , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/analysis , Jejunum , Lymphocyte Activation , Male , Middle Aged , Organ Culture Techniques , Receptors, Interleukin-2 , Statistics, Nonparametric , fas Receptor/analysis
8.
Gut ; 51(2): 177-81, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12117875

ABSTRACT

BACKGROUND AND AIMS: Coeliac disease (CD) is a multifactorial disorder which has an autoimmune component characterised by the occurrence of disease specific autoreactive antibodies against the enzyme tissue transglutaminase (tTG). The aim of this study was to investigate whether binding of antibodies to the enzyme influences tTG activity. METHODS: tTG activity was assayed in the presence of immunoglobulin A (IgA) and immunoglobulin G (IgG) purified from the serum of coeliac patients, CUB 7402 (an anti-tTG mouse monoclonal antibody), and human anti-tTG monoclonal antibodies derived from both intestinal lymphocytes from three patients with CD and from peripheral blood lymphocytes from healthy subjects. For our studies we used calcium treated and untreated recombinant human tTG. Furthermore, the effects of antibodies were determined by immunohistochemical detection of tTG activity in sections of human umbilical cord. RESULTS: IgG and IgA from CD patients inhibited tTG activity in vitro in a dose dependent manner, with a different rate of inhibition among patients. The monoclonal antibody CUB 7402 and human monoclonal antibodies displayed a dose dependent inhibitory effect towards the catalytic activity of the enzyme, both in vitro and in situ. Preincubation of tTG with CaCl(2) caused loss of the inhibitory effect due to CUB 7402 but not that caused by human monoclonal antibodies. CONCLUSIONS: Purified CD IgA, IgG, as well as human anti-tTG monoclonal antibodies inhibited the enzymatic activity of human tTG both in vitro and in situ.


Subject(s)
Antibodies, Monoclonal/pharmacology , Celiac Disease/enzymology , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/immunology , Transglutaminases/immunology , Animals , Calcium Chloride/pharmacology , Celiac Disease/immunology , Cell Line , Cells, Cultured , Dogs , GTP-Binding Proteins/analysis , Humans , Immunoglobulin A/pharmacology , Immunoglobulin G/pharmacology , Immunohistochemistry/methods , Mice , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins/pharmacology , Transglutaminases/analysis , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism , Umbilical Cord/enzymology
9.
Gut ; 50(2): 186-90, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11788557

ABSTRACT

BACKGROUND: Coeliac disease (CD) is caused by a T helper cell type 1 (Th1) response in the small intestinal mucosa to dietary gluten. Paradoxically, interleukin (IL)-12, the major Th1 inducing factor, is undetectable in the mucosa of active CD. IL-18 is a recently described cytokine capable of promoting T cell interferon (IFN)-gamma production and facilitating Th1 cell polarisation. AIM: To examine expression of IL-18 and IL-18-associated Th1 proteins in CD. METHODS: IL-18 and IFN-gamma RNA transcripts were determined by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). IL-18 and caspase-1 protein expression were assessed by western blotting. Caspase-1 activity was determined using a commercially available assay. RNA transcripts for the IL-18 receptor subunits, IL-1 receptor related protein (IL-1 Rrp) and accessory protein-like subunit (AcPL), and IL-18 induced Th1 specific T box transcription factor (T-bet) were measured by RT-PCR and Southern blotting. RESULTS: IL-18 RNA transcripts were found in all mucosal samples analysed, with no difference between CD patients and controls. By western blot analysis, a large protein of approximately 24 kDa, corresponding to the immature IL-18, was detected in all mucosal samples from CD patients and controls. In contrast, mature IL-18 was only seen in CD patients. Immunoreactivity corresponding to both immature and mature caspase-1 was present in both CD and control samples. Tissue homogenates from CD patients and controls expressed similar levels of caspase-1 activity. IL-1Rrp and AcPL were seen in all samples but were expressed at greater levels in the mucosa of CD patients. T-bet was also upregulated in CD. CONCLUSIONS: Active IL-18 is expressed in CD as well as other markers of Th1 polarisation.


Subject(s)
Celiac Disease/immunology , Interleukin-18/metabolism , Receptors, Interleukin , Th1 Cells/metabolism , Adolescent , Adult , Biomarkers/analysis , Caspase 1/metabolism , Celiac Disease/metabolism , Child , Child, Preschool , DNA, Complementary/metabolism , Humans , Infant , Interferon-gamma/metabolism , Interleukin-18 Receptor alpha Subunit , Interleukin-18 Receptor beta Subunit , Intestinal Mucosa/metabolism , Middle Aged , Proteins/metabolism , RNA/metabolism , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-18 , T-Box Domain Proteins , Transcription Factors/metabolism , Up-Regulation
10.
Scand J Immunol ; 53(3): 290-5, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11251887

ABSTRACT

The mucosal lesion present in coeliac disease is an immune-mediated injury triggered by gliadin and restricted by a particular assortment of major histocompatibility complex genes. In view of this, an immunomodulatory approach that induces tolerance to this antigen appears to be a possible alternative to a strict gluten-free diet in treating coeliac disease. We have shown that intranasal administration of multiple doses of whole gliadin is required to specifically inhibit T helper 1-like T-cell reactivity in BALB/c mice immunized parenterally with whole gliadin. However, T-cell activation to multiple antigens, as a consequence of the chemical complexity shown by the antigen gliadin, could hamper efforts to identify single component(s) useful for tolerance induction. In this study, gliadin fractions were purified and administered intranasally to study their ability to induce tolerance to whole gliadin in our animal model. We found that the alpha fraction was particularly effective in downregulating both the in vitro gliadin-specific T-cell proliferation and interferon-gamma production to whole gliadin. In particular, a purified alpha-gliadin was able to suppress the immune response to the entire gliadin mixture. These results demonstrate how an immune response to a complex antigen may be controlled by treatment with a purified component and specifically indicate alpha-gliadin to be a good candidate for further identification of short peptides to be used as tolerogens in this model.


Subject(s)
Gliadin/administration & dosage , Gliadin/immunology , Administration, Intranasal , Animals , Celiac Disease/immunology , Celiac Disease/therapy , Disease Models, Animal , Female , Gliadin/isolation & purification , Humans , Immune Tolerance , Immunity, Mucosal , In Vitro Techniques , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology
11.
Am J Gastroenterol ; 96(1): 150-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11197245

ABSTRACT

OBJECTIVES: Celiac disease (CD) is an under-diagnosed but extremely frequent disease, triggered by the ingestion of gliadin. The pathogenic mechanisms of CD are still poorly understood, but intraepithelial lymphocytes are considered to have a key role. We intended to define the subsets of T lymphocytes migrating upon gliadin challenge in organ cultures of treated celiac patients and establish the type of factor(s) driving such an infiltration. METHODS: Duodenum biopsies from 10 treated celiacs and 7 controls were cultured in vitro with/without gliadin digest (1 mg/ml) or interleukin (IL)-15 (10 ng/ml). In 7 treated celiacs IL-7, IL-4, and IL-2 were similarly tested. Intraepithelial CD3, CD8, TCR-gammadelta, and CD94 were detected by immunohistochemistry and numbered per mm epithelium. RESULTS: IL-15 but not IL-7, IL-4, or IL-2 induced intraepithelial increase of CD3+ and CD8+ cells in celiac and control intestine (p < 0.001 vs cultures with medium). IL-15 induced increases in the number of intraepithelial TCR- gammadelta+ and CD94+ cells only in celiacs (p < 0.001). IL-7 was also effective in increasing intraepithelial TCR-gammadelta+ (but not CD94+) cells in celiac biopsies (p < 0.001). Gliadin induced intraepithelial migration of CD3+, CD8+ (p < 0.001), and CD94+ (p < 0.05) cells in celiacs, but not in controls. CONCLUSIONS: The results we describe in this report indicate that IL-15 might have a key role in modulating and driving intraepithelial infiltration and ultimately in the pathogenesis of CD.


Subject(s)
Antigens, CD/immunology , Celiac Disease/immunology , Cell Movement/immunology , Interleukin-15/immunology , Lectins, C-Type , Lymphocytes/immunology , Membrane Glycoproteins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Adolescent , Adult , Biopsy, Needle , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/immunology , Celiac Disease/diet therapy , Celiac Disease/pathology , Culture Techniques , Female , Humans , Immunohistochemistry , Interleukin-15/pharmacology , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily D , Receptors, Antigen, T-Cell, gamma-delta/drug effects , Reference Values , Sensitivity and Specificity
12.
Gut ; 48(3): 418-24, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171836

ABSTRACT

BACKGROUND: Villus atrophy is the most distinctive sign of untreated coeliac disease (CD) and epithelial apoptosis is considered to be involved in this stage of the coeliac lesion. The extent of villus atrophy is, however, not homogeneous and patients with patchy or mild lesions have been described. AIMS: To address: (a) the degree of "patchiness" in untreated CD patients; and (b) to clarify if apoptosis, and eventually which trigger drives it, causes epithelial damage. PATIENTS: Twenty of 40 untreated, 14 treated coeliac patients, and 15 controls received five or more multiple duodenal biopsies; the remaining 20 untreated CD patients had no more than three biopsies. METHODS: All biopsies were analysed to monitor the presence of a "flat" mucosa. Biopsies of 14 untreated, 10 treated coeliacs, and seven controls were cultured with or without gliadin. DNA fragmentation was studied by terminal deoxynucleotidyl transferase (TdT) mediated dUTP digoxigenin nick end labelling (TUNEL), and FAS and Ki67 expression by immunohistochemistry. Antiendomysium antibodies (EMA) were surveyed in biopsy culture supernatants. RESULTS: A pattern of patchy duodenal lesions was observed in all untreated CD patients biopsied up to five times. High enterocyte FAS expression, and a high number of TUNEL+ and Ki67+ enterocytes were detected in areas with villus atrophy but not in those with a normal morphology (p<0.001). Conversely, EMA in culture supernatants and signs of immunological activation were present in all untreated CD biopsies. In vitro gliadin challenge increased the number of TUNEL+ and Ki67+ enterocytes (p<0.001 v cultures with medium alone) only in "flat" biopsies. Neutralising anti-FAS monoclonal antibodies were found to control gliadin induced enterocyte apoptosis (p>0.01) while agonist anti-FAS monoclonal antibody increased it (p<0.001). CONCLUSIONS: Patchy lesions are observed in untreated CD mucosa and epithelial FAS engagement is a key trigger in driving villus atrophy in CD.


Subject(s)
Apoptosis/immunology , Celiac Disease/physiopathology , Enterocytes/physiology , fas Receptor/physiology , Adolescent , Adult , Antibodies, Monoclonal/physiology , Autoantibodies/physiology , Biopsy , Celiac Disease/metabolism , Cells, Cultured , Child , Child, Preschool , Gliadin/pharmacology , Humans , In Situ Nick-End Labeling , Intestinal Mucosa/physiopathology , Ki-67 Antigen/metabolism , Middle Aged
13.
Gastroenterology ; 119(4): 996-1006, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11040186

ABSTRACT

BACKGROUND & AIMS: Villous atrophy and crypt proliferation are key epithelial features of untreated celiac disease. We tried to define whether cytokines such as interleukin (IL)-15, IL-2, IL-4, and IL-7, which share chains of their receptors, could influence the epithelial modifications. METHODS: Duodenal biopsy specimens (14 treated and 13 untreated celiac patients, 7 controls) were cultured in vitro for 24 hours with or without gliadin (1 mg/mL), IL-15, IL-7, IL-4, or IL-2 (10 ng/mL). Tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma were also used in some specimens of untreated celiacs. Epithelial expression of Ki67, FAS, and transferrin receptor (TFR) was detected by immunohistochemistry, and apoptosis by TUNEL technique (percentage of positive enterocytes). IL-15-positive cells were detected by immunohistochemistry in celiac disease and control biopsy specimens; presence of IL-15 was also determined by semiquantitative polymerase chain reaction. RESULTS: Only IL-15 induced enterocyte expression of Ki67, TFR, and FAS in treated celiac (P<0.01 vs. medium) and enterocyte apoptosis in untreated celiac disease specimens. Anti-IL-15 monoclonal antibodies neutralized gliadin-induced enterocyte TFR and FAS expression in treated celiac and enterocyte apoptosis in untreated celiac disease specimens (P<0.05 vs. gliadin). IL-15-positive cells were increased in untreated celiacs (P<0.001 vs. treated celiacs and controls). CONCLUSIONS: IL-15 is involved in the modulation of epithelial changes in celiac disease, indicating that this cytokine has an unforeseen role in the pathologic manifestations of celiac disease.


Subject(s)
Celiac Disease/immunology , Celiac Disease/pathology , Cytokines/pharmacology , Interleukin-15/genetics , Interleukin-15/pharmacology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Adolescent , Adult , Antibodies, Monoclonal/pharmacology , Biopsy , Celiac Disease/genetics , DNA Fragmentation , Duodenum , Female , Gliadin/pharmacology , Humans , Interferon-gamma/pharmacology , Interleukin-15/analysis , Interleukin-2/pharmacology , Interleukin-4/pharmacology , Interleukin-7/pharmacology , Intestinal Mucosa/drug effects , Jejunum , Male , Middle Aged , Organ Culture Techniques , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
14.
Pediatr Res ; 47(3): 344-50, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10709733

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator gene and characteristically leads to prominent lung and pancreatic malfunctions. Although an inflammatory reaction is normally observed in the CF airways, no studies have been performed to establish whether a chronic inflammatory response is also present in the CF intestine. We have investigated whether immunologic alterations and signs of inflammation are observed in CF small intestine. Fourteen CF, 20 negative, and four disease controls underwent duodenal endoscopy for diagnostic purposes. Two CF patients were rebiopsied, one after 3 mo of an elemental diet and the other after 2 wk of pancreatic enzyme withdrawal. In three CF and 10 controls, in vitro small intestine organ cultures were also performed. Expression of ICAM-1, IL-2 receptor, IL-2, IFN-gamma, CD80, and transferrin receptor was studied by immunohistochemistry before and after in vitro organ culture. In CF small intestine, an increased number of lamina propria mononuclear cells express ICAM-1 [mean 114 (SD 82.8), p < 0.001 versus controls], CD25 [20.2 (18.7), p < 0.01], IL-2 [23.6 (13.7), p < 0.05], and IFN-gamma [19 (15.9), p < 0.05], whereas villus enterocytes highly express transferrin receptor. Reduced expression of immunologic markers was observed after 24 h of in vitro culture in all three CF patients as well as in the patient kept on elemental diet for 3 mo. These results indicate that chronic inflammation is observed in CF duodenum and suggest that the perturbation of local mucosal immune response may contribute to the overall clinical picture in CF patients.


Subject(s)
Cystic Fibrosis/pathology , Intestine, Small/pathology , Adolescent , Adult , Child , Child, Preschool , Chronic Disease , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Female , Humans , Immunohistochemistry , Intestine, Small/immunology , Intestine, Small/metabolism , Male
16.
Scand J Immunol ; 50(2): 177-82, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10447922

ABSTRACT

The mucosal lesion in coeliac disease (CD) represents an immunologically mediated injury triggered by gliadin and is restricted by a particular assortment of major histocompatibility complex (MHC) class II genes. Therefore, immunomodulatory strategies to tolerize gliadin-specific, class II-restricted T-cell responses could represent an alternative to current treatments of CD, which are based on a gluten-free diet. In this study, BALB/c mice derived from a gluten-free diet colony were tolerized by either intranasal (i.n.) or intravenous (i.v.) administration of single or multiple doses of gliadin. While a single dose failed to induce tolerance, a significant decrease in gliadin-specific T-cell proliferation was detected (P < 0.001) after multiple i.n. or i.v. administrations. No significant difference in antibody titre was detected for antigen-specific immunoglobulin G (IgG) or the IgG1 subclass, but a lower IgG2a-specific titre was observed. Both interferon-gamma (IFN-gamma) and interleukin (IL)-2 expression, measured by enzyme-linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR), were reduced on antigen administration, both i.v. and i.n. Neither regimen showed a regulatory effect on IL-4 production. As T helper 1 (Th1) cytokines seem to be important in the pathogenesis of CD, our data therefore highlight the potential of i.n. and i.v. routes for the design of useful immunomodulatory strategies for CD.


Subject(s)
Gliadin/immunology , Administration, Intranasal , Animals , Cell Division , Cells, Cultured , Down-Regulation , Female , Gliadin/administration & dosage , Immunoglobulin G/immunology , Injections, Intravenous , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred BALB C , T-Lymphocytes/cytology , T-Lymphocytes/immunology
17.
Dig Dis Sci ; 44(5): 960-5, 1999 May.
Article in English | MEDLINE | ID: mdl-10235604

ABSTRACT

During embryogenesis, two different transmembrane receptors, Ret and Ednrb, together with their ligands, respective, GDNF and endothelin-3, are involved in the migration and differentiation of enteric ganglion cells, sympathetic neurons and melanocytes from the neural crest. Mutations in these genes have been found in a number of human and murine neurocristopathies, including Hirschsprung's disease. RET and GDNF knockouts suggest that they are involved in a correct autonomic nervous system formation. The aim of this study is the evaluation of the autonomic nervous system in patients with Hirschsprung's disease. Seventeen children (mean age: 8.6 years) with Hirschsprung's disease and 19 age- and sex-matched control children (mean age: 9.9 years) underwent pupillary and cardiovascular testing of sympathetic adrenergic and cholinergic function and cardiovagal cholinergic function. Seven of 17 patients with Hirschsprung's disease were affected by autonomic dysfunction. Three of seven patients had evidence of sympathetic denervation, two showed a parasympathetic dysfunction, whereas the remaining two had dysfunction of both sympathetic and parasympathetic tests. Our data in a small number of patients with Hirschsprung's disease show that a subset of these patients exhibits measurable autonomic dysfunction. A RET mutation has been found in one of them. As for the absence of the enteric ganglion cells, autonomic dysfunction in these subjects seems to be polygenic.


Subject(s)
Autonomic Nervous System/physiopathology , Hirschsprung Disease/physiopathology , Adolescent , Child , Child, Preschool , DNA Mutational Analysis , Female , Ganglia, Sensory/cytology , Hirschsprung Disease/genetics , Humans , Intestines/cytology , Male
18.
Ital J Gastroenterol Hepatol ; 31(8): 773-80, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10730570

ABSTRACT

Coeliac disease is a permanent intolerance to wheat gliadins and related prolamines. Patients who have an obvious malabsorption syndrome form only a small minority of the total number of people with coeliac disease. There are, in fact, no pathognomonic clinical features, and the condition is defined and diagnosed by the presence of pathological changes in the small bowel mucosa related to the presence of toxic prolamines. Susceptibility to coeliac disease is determined to a significant extent by genetic factors. A large part of the genetic susceptibility maps to the HLA region on chromosome 6, as approximately 95% of coeliac disease patients carry an almost identical HLA DQ2/heterodimer; a role of non-HLA genes has also been postulated. From a pathogenetic point of view, most evidence supports the notion of a DQ-restricted gluten-specific Th1 response in the lamina propria; nonetheless, it is possible that, in coeliac subjects, gluten, prior to T cell activation, could exert a direct toxic effect leading to the production of proinflammatory signals.


Subject(s)
Celiac Disease , Celiac Disease/diagnosis , Celiac Disease/genetics , Celiac Disease/physiopathology , Celiac Disease/therapy , Humans
20.
Gut ; 43(4): 484-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9824574

ABSTRACT

BACKGROUND: CD3 and gamma delta cells in the rectal mucosa increase after local instillation of gluten in children with coeliac disease and in half of their siblings. Aim- To establish an in vitro system for assessing immunological changes induced by gluten in the rectum. PATIENTS AND METHODS: Rectal biopsy specimens obtained from 13 treated coeliac children, nine of their siblings, and nine controls were cultured in vitro with a peptic-tryptic digest of gliadin or ovalbumin. CD3 and CD25 cells were counted, and the expression of adhesion molecules evaluated. RESULTS: In the lamina propria of coeliac biopsy samples cultured with gliadin, but not in those from controls, the expression of vascular cell adhesion molecule 1 (VCAM-1) was enhanced, and the number of CD25 cells was significantly higher than in those cultured in medium alone; the density of intraepithelial CD3 cells was also significantly higher. No differences were noted in coeliac biopsy specimens cultured with ovalbumin. A discriminant analysis allowed correct classification of all controls and all coeliacs but one, but three of nine siblings were allocated to the coeliac group. CONCLUSIONS: Our data confirm that gliadin is able to activate cell mediated immunity in the rectal mucosa in coeliac patients and in a subset of their first degree relatives.


Subject(s)
Celiac Disease/immunology , Gastric Mucosa/immunology , Gliadin/immunology , Adolescent , Adult , CD3 Complex/metabolism , Celiac Disease/genetics , Child , Female , Humans , Immunity, Cellular , Immunity, Mucosal , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Male , Pedigree , Receptors, Interleukin-2/metabolism , Sensitivity and Specificity , Staining and Labeling , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...